EP2109624B1 - Procede de diagnostic de maladies par depistage de l'hepcidine dans des tissus, du sang ou des liquides organiques humains ou animaux, anticorps monoclonaux specifiques de l'hepcidine humaine et leurs utilisations associees - Google Patents

Procede de diagnostic de maladies par depistage de l'hepcidine dans des tissus, du sang ou des liquides organiques humains ou animaux, anticorps monoclonaux specifiques de l'hepcidine humaine et leurs utilisations associees Download PDF

Info

Publication number
EP2109624B1
EP2109624B1 EP07723512.5A EP07723512A EP2109624B1 EP 2109624 B1 EP2109624 B1 EP 2109624B1 EP 07723512 A EP07723512 A EP 07723512A EP 2109624 B1 EP2109624 B1 EP 2109624B1
Authority
EP
European Patent Office
Prior art keywords
hepcidin
antibody
seq
amino acid
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP07723512.5A
Other languages
German (de)
English (en)
Other versions
EP2109624A1 (fr
Inventor
Hasan Kulaksiz
Cyril E. Geacintov
Alfred Janetzko
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DRG International Inc
Original Assignee
DRG International Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39645313&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2109624(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by DRG International Inc filed Critical DRG International Inc
Publication of EP2109624A1 publication Critical patent/EP2109624A1/fr
Application granted granted Critical
Publication of EP2109624B1 publication Critical patent/EP2109624B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention concerns methods and kits for diagnosing a disease condition characterized by non-physiological levels of hepcidin protein.
  • the present invention further concerns monoclonal antibodies that interact specifically with the mature form of human hepcidin (hepcidin-25). Multiple forms of these isolated antibodies are disclosed and exemplified herein.
  • the present invention further concerns methods of treating any disease condition within a patient that is characterized by such non-physiological levels of hepcidin protein.
  • Iron is an essential trace element that is required for growth and development of all living organisms. It is indispensable for DNA synthesis and is an essential component of many proteins and enzymes including haemoglobin and myoglobin, the cytochromes, NADH dehydrogenase, lipooxygenases, phosphatases, superoxide dismutase, ribonucleotide reductase, and fatty acid desaturases.
  • Iron can also be toxic when present in excess because of its ability to generate reactive oxygen species. This dual nature imposed a very tight regulation of the iron concentration in the body. Disturbances in iron metabolism are implicated in a number of significant human diseases, including anemia of chronic diseases, anemia of inflammation, or the iron overload disease hemochromatosis.
  • iron absorption occurs predominantly in the duodenum and upper jejunum, and systemic iron homeostasis is regulated at the level of intestinal absorption and this is the only mechanism by which iron stores are physiologically controlled ( Philpott, Hepatology 35:993-1001 2002 ).
  • iron is bound to circulating transferrin and delivered to tissues throughout the body.
  • the liver is the major site of iron storage.
  • transferrin-bound iron is taken into the hepatocytes by receptor-mediated endocytosis via the classical transferrin receptor (TfR1) ( Collawn et al.
  • TfR2 homologous transferrin receptor 2
  • TfR2 homologous transferrin receptor 2
  • the extracellular domain of this protein is 45% identical to the corresponding portion of TfR1 (Id.).
  • TfR2 can also bind diferric transferrin and facilitate the uptake of iron. Mutations in TfR2 have been associated with certain forms of hemochromatosis demonstrating the important role for TfR2 in iron homeostasis ( Philpott, Hepatology 35:993-1001, 2002 ; Camasehella et al., Nat. Genet.
  • TfR2 is predominantly expressed in the liver ( Fleming et al., Proc. Natl. Acad. Sci. USA 97: 2214-2219, 2000 ; Subramaniam et al., Cell Biochem. Biophys. 36:235-239, 2002 ), and is localized in the basolateral membrane domain of hepatocytes. ( Merle et al., Histochem. Cell. Biol., 2006 .)
  • Hepcidin is a recently discovered peptide hormone ( Park et al., J. Biol. Chem. 276:7806-7810, 2001 ; Krause et al., FEBS Letter 480:147-150, 2000 ), which is the key regulator of systemic iron homeostasis. Hepcidin is predominantly produced in the liver ( Park et al. J. Biol. Chem. 276:7806-7810, 2001 ; Kulaksiz et al. GUT 53:735-43, 2004 ), circulates in plasma and is excreted in urine ( Kulaksiz et al., J. Endocrinol. 184, 2005 ).
  • hepcidin is encoded by a small three-exon gene as a preprohepcidin with a characteristic signal sequence and a furin cleavage site preceding the mature hepcidin peptide.
  • the active form of the peptide is a 25 amino acid ß-sheet hairpin stabilized by four disulfide bonds. It is synthesized as a preprohepcidin of 84 amino acids.
  • the signal peptide is cleaved leading to the 60 amino acids prohepcidin, which is further processed giving rise to the 25 amino acids hepcidin. In human urine, the predominant form is the 25 amino acid peptide, although shorter peptides with 20 and 22 amino acids are also detectable.
  • Hepcidin-20, hepcidin-22 and hepcidin-25 are, for example, referred to collectively as the mature form of hepcidin.
  • hepcidin The involvement of hepcidin in iron metabolism was suggested by the observation that hepcidin synthesis is induced by dietary iron ( Pigeon et al. J. Biol. Chem. 276:7811-7819, 2001 ). The specific role of hepcidin was then examined by assessing the effects of its deficiency or excess in transgenic mouse models. Hepcidin expression is abolished in mice exhibiting iron overload due to targeted disruption of the upstream stimulatory factor 2 (Usf2) gene, resembling the same phenotype as found in hfe-/- mice ( Nicolas et al., Proc. Natl. Acad. Sci. USA 98:8780-8785, 2001 ).
  • Usf2 upstream stimulatory factor 2
  • hepcidin is a central regulator of iron homeostasis.
  • liver hepcidin expression is decreased in the hfe knockout mouse ( Ahmad et al., Blood Cells Mol. Dis. 29, 2002 ), and mutations in the hepcidin peptide are associated with severe juvenile hemochromatosis ( Roetto et al., Nat. Genet. 33, 2003 ), providing new perspectives in our understanding of the molecular pathogenesis of iron overload.
  • hepcidin inhibits cellular efflux of iron by binding to ferroportin ( Nemeth et al. Science 306: 2090-2093, 2004 ), the only known mammalian iron exporter, which is expressed by enterocytes, macrophages and hepatocytes.
  • ferroportin the only known mammalian iron exporter, which is expressed by enterocytes, macrophages and hepatocytes.
  • the binding of hepcidin causes ferroportin to be internalized and degraded, and the loss of ferroportin from cell membrane ablates cellular iron export.
  • the direct hepcidin-ferroportin interaction allows an adaptive response from the body in situations that alter normal iron homeostasis (hypoxia, anemia, iron deficiency, iron overload, and inflammation).
  • hepcidin production is homeostatically regulated by anemia and hypoxemia ( Nicolas et al., J. Clin. Invest. 110, 2002 ).
  • oxygen delivery is inadequate, the homeostatic response is to produce more erythrocytes.
  • hepcidin levels decrease, its inhibitory effects diminish, and more iron is made available from the diet and from the storage pool in macrophages and hepatocytes.
  • Hepcidin as an iron-regulatory hormone, constitutes an important link between host defense, inflammation and iron metabolism. Hepcidin is structurally similar to cysteine rich, cationic, antimicrobial peptides, including the defensins and some cathelicidins. In vitro, human hepcidin exerts antimicrobial and antifungal activities ( Park et al., J. Biol. Chem. 276:7806-7810, 2001 ; Krause et al., FEBS Letter 480:147-150, 2000 ). Its synthesis is markedly induced by infection and inflammation ( Pigeon et al., J. Biol. Chem.
  • the cytokine IL-6 is apparently the key inducer of hepcidin synthesis during inflammation ( Nemeth et al., J. Clin. Invest. 113, 2004 ) and anti-IL-6 antibodies block the induction of hepcidin mRNA in human hepatocyte cell lines treated with supernatants of LPS- or peptidoglycan-stimulated macrophages.
  • anti-IL-6 antibodies block the induction of hepcidin mRNA in human hepatocyte cell lines treated with supernatants of LPS- or peptidoglycan-stimulated macrophages.
  • normal mice show a marked decrease in serum iron ( Nicolas et al., J. Clin. Invest. 110, 2002 ; Nemeth et al., J. Clin. Invest. 113, 2004 ).
  • hepcidin in iron homeostasis and its disorders suggests that its assay in blood or urine could prove useful for the diagnosis and monitoring of iron disorders. Furthermore, hepcidin could be a marker for disease activity of chronic inflammatory diseases such as, for example, chronic polyarthritis or Crohn's disease, or ulcerative colitis Currently, there is no stable and validated Enzyme-Linked ImmunoSorbent Assays (i.e.
  • hepcidin-25 is the bioactive form ( Nemeth et al., Science 2004, 306: 2090-2093 ; Krause et al., FEBS Letters 480, 147-150, 2000 ; Park et al., Journal of Biological Chemistry 276, 7806-7810, 2001 ; Rivera et al., Blood 106, 2196-2199, 2005 ), and may play a critical role in the pathogenesis of diseases associated with a disturbance of iron metabolism ( Pietrangelo A., N. Engl. J. Med 2004, 350: 2383-2390 ; Ganz T., Curr Top. Microbiol. Immunol., 2006; 306: 183-198 ; Hugman A, Clin.
  • the invention addresses drawbacks of the prior art by providing, in one aspect, sensitive methods and kits for diagnosing a disease condition characterized by non-physiological levels of hepcidin protein.
  • a portion of the present invention relates to an antibody or antibody fragment which interacts with a specific region of a mature form of human hepcidin-25, namely amino acids 74-81 of hepcidin-25, represented as His Arg Ser Lys Cys Gly Met Cys (SEQ ID NO:3).
  • the present invention further relates to an antibody raised against a specific region of mature human hepcidin-25 which comprises the region from amino acid 74-81, wherein amino acid residue 78 is Xaa (His Arg Ser Lys Xaa Gly Met Cys (SEQ ID NO:4]) , where Xaa is any amino acid selected from the group consisting of L or D stereoisomeric forms of the 20 common amino acids.
  • amino acid residue 78 is alpha aminobutryic acid (His Arg Ser Lys Abu Gly Met Cys [SEQ ID NO:5]).
  • the present invention relates to a monoclonal antibody or fragment thereof which interacts with a specific region of human hepcidin-25 (again, amino acids 74-81 of hepcidin-25, represented as His Arg Ser Lys Cys Gly Met Cys [SEQ ID NO:3]). Therefore, the present invention relates to a monoclonal antibody raised against a specific region of human hepcidin-25 which comprises the region from amino acid 74-81, wherein amino acid residue 78 is Xaa (His Arg Ser Lys Xaa Gly Met Cys [SEQ ID NO:4]), as disclosed herein.
  • an exemplified peptide susbstitutes alpha aminobutryic acid for cysteine (His Arg Ser Lys Abu Gly Met Cys [SEQ ID NO:5]), which was used as an immunogen to generate the exemplified monoclonal antibodies described herein.
  • the present invention also relates to a hybridoma capable of producing a monoclonal antibody of the present invention.
  • Particular hybridomas of the present invention are hybridomas which produce monoclonal antibodies mHK(5), mHK(8/1), mHK(8/2), mHK(8/3) and mHK(9), respectively.
  • the antibodies of the present invention may also be in the form of a polyclonal sera raised against this specific region of human hepcidin-25, especially polyclonal sera raised against this core sequence comprising at least amino acid residues 74-81 (His Arg Ser Lys Cys Gly Met Cys [SEQ ID NO:3]), His Arg Ser Lys Xaa Gly Met Cys [SEQ ID NO:4], and/or His Arg Ser Lys Abu Gly Met Cys [SEQ ID NO:5]).
  • the present invention further relates to a peptide which comprises the amino acid sequence shown as His Arg Ser Lys Cys Gly Met Cys (SEQ ID NO:3).
  • the present invention also relates to a peptide which comprises the amino acid sequence shown as His Arg Ser Lys Xaa Gly Met Cys (SEQ ID NO:4), where Xaa is any amino acid selected from the group consisting of L or D stereoisomeric forms of the 20 common amino acids.
  • the present invention also relates to a peptide which comprises the amino acid sequence shown as His Arg Ser Lys Abu Gly Met Cys (SEQ ID NO:5), where amino acid residue 78 is alpha aminobutryic acid.
  • the present invention also relates to a peptide which consists of the amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:4 and SEQ ID NO:5.
  • the present invention also relates to methods of generating antibodies specific for mature human hepcidin which comprises immunizing a non-human animal with a C-terminal hepcidin peptide which comprises the amino acid sequence as shown in of SEQ ID NO:3, SEQ ID NO:4, and/or SEQ ID NO:5.
  • the present invention further relates to methods and assays for quantitatively determining hepcidin levels, which comprises obtaining a tissue or fluid sample from a subject; contracting the sample with an antibody or fragment thereof that specifically binds to a peptide selected from the group consisting of SEQ ID NO:3, SEQ ID NO:4, and/or SEQ ID NO:5., and quantifying the hepcidin level in the sample; wherein the non-physiological level of hepcidin is indicative of the disease condition.
  • kits for quantitatively determining hepcidin levels which comprises obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to a peptide selected from the group consisting of SEQ ID NO:3, SEQ ID NO:4, and/or SEQ ID NO:5, and quantifying the hepcidin level in the sample; wherein the non- physiological level of hepcidin is indicative of the disease condition.
  • the present invention also relates to methods, assays and kits for quantitatively determining prohepcidin and/or mature hepcidin levels, which comprises obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to one or more epitopes of human hepcidin precursor amino acid sequence between and including amino acids 74-81 (i.e., SEQ ID NO:3), and quantifying the hepcidin level in the sample; wherein the non- physiological level of hepcidin is indicative of the disease condition.
  • the present invention also relates to methods, assays and kits for quantitatively determining hepcidin levels, which comprises obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to one or more epitopes of human hepcidin precursor amino acid sequence between and including amino acids 60-84 (i.e., SEQ ID NO:2), and quantifying the hepcidin level in the sample; wherein the non- physiological level of hepcidin is indicative of the disease condition.
  • the present invention concerns hepcidin regulation of iron uptake by mammalian cells and the use of hepcidin and/or hepcidin specific antibodies in the diagnosis of diseases involving disturbances of iron metabolism.
  • the diagnostic detection kits of the present invention can be particularly useful in screening the overall population of either humans or animals and identifying those subjects who have these diseases.
  • one aspect of the invention relates to a method for diagnosing a disease condition characterized by non-physiological levels of hepcidin, comprising obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to a polypeptide from the mid-portion (amino acids 20 to 50, SEQ. ID. NO. 1) or C-terminus of hepcidin (amino acids 60 to 84, SEQ. ID. NO.
  • hepcidin protein such as a region of the human hepcidin protein comprising amino acids 74-81 (His Arg Ser Lys Cys Gly Met Cys [SEQ ID NO:3]), and quantifying the hepcidin level using an assay based on binding of the antibody and the polypeptide; wherein the non-physiological level of hepcidin is indicative of the disease condition.
  • sensitive diagnostic methods and kits were established enabling the detection of pro-hepcidin in human plasma.
  • hepcidin e.g., HEPCIDIN-25, HEPCIDIN-22, and/or HEPCIDIN-20; see Figure 8
  • a hepcidin antibody and diagnostic methods e.g., HEPCIDIN-25, HEPCIDIN-22, and/or HEPCIDIN-20; again, see Figure 8
  • kits can be used for the determination of hepcidin as a parameter for the progress of the diseases mentioned above during and after therapy.
  • This invention further provides the demonstration that a hepcidin protein in subjects of these disorders are present in human or animal tissue, blood and body fluids in concentrations greatly exceeding that found in normal humans or animals that are not subjects of these disorders. This is achieved by examining a sample of tissue, blood or body fluid from a patient, and detecting the presence and quantity of hepcidin protein, including but not limited to prohepcidin or any mature form of hepcidin, such as hepcidin-25, or a 22- or 20-amino acid version of mature hepcidin (see Figure 8 ).
  • the detection and quantitative measurement of any hepcidin protein or fragment thereof in tissue, blood or body fluids in accordance with this invention is useful in confirming a clinical diagnosis of the diseases described herein, in affected patients and in following the course of the disease.
  • the invention is also useful in monitoring the disease during and subsequent to a period of treatment with agents that are being tested for their ability to stabilize, decrease or prevent the occurrence of such diseases.
  • Nonphysiological amounts of the hepcidin protein or a fragment thereof can exist in disturbances of iron metabolism, resulting in iron deficiency or overload, such as iron deficiency anemia, genetic and nongenetic iron overload diseases, such as hemosiderosis and hemochromatosis, or secondary hemochromatosis, aceruloplasminemia, hypotransferrinemia, atransferrinemia; iron overload diseases of undetermined origin, for instance in the case of diseases of the biliary system, liver diseases, especially alcoholic liver diseases, nonalcoholic steatohepatitis, and chronic hepatitis B and C infections; diseases of utilization of iron, such as sideroblastic anemia, thalassemia; hematologic diseases, such as leukemia, polyglobulie, macrocytic, microcytic or normocytic anemia, anemia with reticulocytosis, hemolytic anemia; disturbances of the reticuloendothelial system due to infections and diseases; inflammations and infections, including
  • An embodiment of the invention concerns hepcidin specific antibodies, or fragments or variants thereof that, in turn, can be used in immunoassays to detect a hepcidin protein in suspected humans or animals.
  • the hepcidin diagnostic methods and kits can be used in genetic technological approaches, such as for overexpressing or downregulating hepcidin.
  • hepcidin can be used in therapeutic treatment of the diseases described herein, by treating subjects with hepcidin, and agonists or antagonists of hepcidin, including but not limited to monospecific antibodies, especially monospecific antibodies of the present invention which recognize one or more epitopes associated with amino acids 74-81 or human hepcidin (see Figure 8 ).
  • Iron uptake in cells may be modulated by varying the concentration of hepcidin, thus modulating binding between hepcidin and ferroportin.
  • hepcidin, and agonists or antagonists of hepcidin may be useful in the treatment of conditions where there is a disturbance in iron metabolism.
  • such substances may be useful in the treatment of such aforementioned diseases.
  • the therapeutic agent may comprise an antibody as mentioned above, including those discussed in the instant application, as well as antisense therapy (including RNAi, siRNA AND shRNA), or agents which alter blood glucose level, and any other molecule which possesses the ability to modulate hepcidin concentration, acting as either an agonist or antagonist of hepcidin concentration.
  • the current invention concerns the following:
  • the present invention describes that hepcidin regulates iron uptake by mammalian cells and nonphysiological express of hepcidin results in disease involved distribution of iron metabolism.
  • Our preliminary data show that the physiological concentration of pro-hepcidin is in the range of 51.6-153.4 ng/mL (the mean concentration is about 106.2 ng/mL).
  • the mean concentration is about 106.2 ng/mL.
  • nonphysiological concentrations are below or over this range.
  • these data may be modified.
  • Nonphysiological amounts of prohepcidin and/or mature hepcidin protein or fragments thereof are associated with disturbances of iron metabolism, resulting in iron deficiency or overload, such as iron deficiency anemia; genetic and nongenetic iron overload diseases, such as hemosiderosis and hemochromatosis or secondary hemochromatosis, aceruloplasminemia, hypotransferrinemia, atransferrinemia; iron overload diseases of undetermined origin, for instance in the case of diseases of the biliary system, liver diseases, especially alcoholic liver diseases, nonalcoholic steatohepatitis, and chronic hepatitis B and C infections; diseases of utilization of iron, such as sideroblastic anemia, thalassemia; hematologic diseases, such as leukemia, polyglobulie, macrocytic, microcytic or normocytic anemia, anemia with reticulocytosis, hemolytic anemia; disturbances of the reticuloendothelial system due to infections and diseases;
  • iron overload includes cirrhosis of the liver and hepatocellular cancer, diabetes, heart failure, arthritis, and hypogonadism. Zhou et al., Proc. Natl. Acad. Sci., 95, 2492-2497 (1998 ).
  • Other suitable diseases include, without limitation, renal anemia, anemia of chronic diseases, anemia in Crohns disease, anemia in ulcerative colitis, sprue, cholangitis, primary or secondary sclerosing cholangitis, chronic polyarthritis, thalassemia, and iron overload after iron.
  • This discovery has permitted the development of assays for a hepcidin protein and fragments thereof and their subsequent purification with retention of their native configuration and physiological activity.
  • the invention is based, in part, on the discovery that in patients suffering from certain disorders a hepcidin protein is present in tissue, blood and body fluid of a human or animal.
  • the present invention relates to use of the antibodies of the present invention in methods of diagnosis, related assays and kits for such diagnosis, as well as therapeutic intervention of various disease states disclosed herein.
  • the present invention solves this need of the art and further discloses novel antibodies which specifically bind to an epitope within the C-terminus of human hepcidin protein.
  • These antibodies are advantageous compared to the prior art in at least two respects.
  • the antibodies of the instant invention are suitable for diagnostic tests such as ELISA, exhibiting a greatly heightened sensitivity to the detection of hepcidin, and second, they can detect and bind the 20-25 amino acid long mature hepcidin.
  • the antibodies can be used to block hepcidin and inhibit its activity, and, accordingly, may be used as a therapeutic agent.
  • the present invention relates in part to an antibody or antibody fragment which interacts with a specific region of a mature form of human hepcidin-25, namely amino acids 74-81 of hepcidin-25, represented as His Arg Ser Lys Cys Gly Met Cys (SEQ ID NO:3), which is also present in pro-hepcidin (see Figure 8 ).
  • an antibody raised against a specific region of mature human hepcidin-25 which comprises the region from amino acid 74-81, wherein amino acid residue 78 is Xaa (His Arg Ser Lys Xaa Gly Met Cys [SEQ ID NO:4]), where Xaa is any amino acid selected from the group consisting of L or D stereoisomeric forms of the 20 common amino acids.
  • a specific embodiment, as disclosed and discussed herein, is an antibody raised against an immieux representing amino acids 74-81 of human hepcidin, whereby amino acid residue 78 is alpha aminobutryic acid (His Arg Ser Lys Abu Gly Met Cys [SEQ ID NO:5]).
  • an antibody of the present invention is a monoclonal antibody or fragment thereof which interacts with a specific region of human hepcidin-25 (e.g., including but not limited to amino acids 74-81 of human hepcidin, represented as His Arg Ser Lys Cys Gly Met Cys [SEQ ID NO:3], see FIG. 8 ). Therefore, the present invention relates to a monoclonal antibody raised against a specific region of human hepcidin-25 which comprises the region from amino acid 74-81, wherein amino acid residue 78 is Xaa (His Arg Ser Lys Xaa Gly Met Cys [SEQ ID No:4]), as disclosed herein.
  • an exemplified peptide susbstitutes alpha aminobutryic acid for cysteine (His Arg Ser Lys Abu Gly Met Cys [SEQ ID NO:5]), which was used as an immunogen to generate the exemplified monoclonal antibodies described herein.
  • any hybridoma capable of producing a monoclonal antibody of the present invention are hybridomas which produce monoclonal antibodies mHK(5), mHK(8/1), mHK(8/2), mHK(8/3) and mHK(9), respectively.
  • the antibodies of the present invention may also be in the form of a polyclonal sera raised against this specific region of human hepcidin-25, especially polyclonal sera raised against this core sequence comprising at least amino acid residues 74-81 (His Arg Ser Lys Cys Gly Met Cys [SEQ ID NO:3]), His Arg Ser Lys Xaa Gly Met Cys [SEQ ID NO:4], and/or His Arg Ser Lys Abu Gly Met Cys [SEQ ID NO:5]).
  • SEQ ID NO: 4 results in antibodies which are, for example, 1) specific to the C-terminal end of hepcidin; 2) highly sensitive for the use in detection and quantification of hepcidin in fluid samples (e.g.
  • ELISA ELISA
  • SEQ ID NO: 4 represents an eight amino acid long peptide corresponding to amino acids 74-81 of the preprohepcidin protein.
  • the hepcidin protein contains cysteine residues which form disulfide bridges with the cysteine residues at positions 70 and 69 respectively.
  • these disulfide bridges contribute to a complicated folded, tertiary loop structure of the C-terminal end of hepcidin.
  • this peptide does not and cannot reflect the epitope reflected by the same eight amino acids as they appear in nature as an essential component of the hepcidin protein.
  • SEQ ID NO: 4 may be found either 1) as an isolated linearized structure, or 2) when "x" is a cysteine (e.g. SEQ ID NO: 3), as a folded structure on account of a possible disulfide bridge formation between cysteine residues at positions 70 and 81.
  • a cysteine e.g. SEQ ID NO: 3
  • Fig. 8 To better understand such a folded structure, reference is made to Fig. 8 and the formation of a cysteine disulfide bridge between positions 6 and 7.
  • Neither the linear or folded structure are present in the natural hepcidin protein and thus are not expected to provide a representative antigen for establishing antibodies specific to this region of the natural hepcidin.
  • cysteine residue e.g.
  • the resulting peptide fragment does not agree with the natural hepcidin protein either in regard to amino acids (i.e. identity) or in regard to structure (i.e. three-dimensional folding), and thus in no manner represents the natural, physiological structure of hepcidin (i.e. that which is expected as necessary for the establishment of effective antibodies).
  • SEQ ID NO: 4 represented, for example, with "x" as alpha aminobutyric acid (i.e. SEQ ID NO: 5), results in the production of a series of monoclonal antibodies (i.e. mHK(5), mHK(8/1), mHK(8/2), mHk(8/3), and mHK(9)) which provide for a more sensitive detection of hepcidin over the prior art (e.g. mHK(9) 8 fold higher sensitivity over the prior art).
  • mHK(9) 8 fold higher sensitivity over the prior art.
  • the prior art provides antibody detection of prohepcidin using antibodies specific to the proregion of hepcidin (i.e. amino acids 25-59 as depicted in Figure 8 ), in particular Kulaksiz et al., Gut, 53, 735-743, 2004 ; US 2004/0096987 A1 ; and US 2004/0096990 A1 provide antibodies EG(1)-HepN and EG(2)-HepN which are specific to amino acids 28-47 of hepcidin. These antibodies, however, only allow for the detection of prohepcidin.
  • the antibodies of the current invention on account of the C-terminal specificity, allow for the detection of both prohepcidin and mature hepcidin.
  • the antibodies of the current invention allow the general use of subtraction assays (e.g. the use of the antibodies of the current invention and the use of the prior art antibodies in parallel experiments and then the subtraction of the results from the prior art antibodies from that of the antibodies of the current invention) to detect and quantify the amount of mature hepcidin in samples.
  • the antibodies of the current invention allow now for the pan reduction or inactivation of hepcidin (i.e. both prohepcidin and mature hepcidin) for the treatment of conditions caused by abnormal iron metabolism.
  • the present invention further relates to a peptide which comprises the amino acid sequence shown as SEQ. ID. NO. 2 or as His Arg Ser Lys Cys Gly Met Cys (SEQ ID NO:3), the amino acid sequence shown as His Arg Ser Lys Xaa Gly Met Cys (SEQ ID NO:4), where Xaa is any amino acid selected from group consisting of L or D stereoisomeric forms of the 20 common amino acids and the amino acid sequence shown as His Arg Ser Lys Abu Gly Met Cys (SEQ ID NO:5), where amino acid residue 78 is alpha aminobutryic acid.
  • the present invention also relates to methods of generating antibodies specific for mature human hepcidin which comprises immunizing a non-human animal with a C-terminal hepcidin peptide which comprises the amino acid sequence as shown in SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, and/or SEQ ID NO:5.
  • the present invention also relates to a peptide which consists of the amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:4 and SEQ ID NO:5.
  • the present invention relates in part to methods and assays for quantitatively determining hepcidin levels, which comprise obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to a peptide selected from the group consisting of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, and/or SEQ ID NO:5, and quantifying the hepcidin level in the sample; wherein the non- physiological level of hepcidin is indicative of the disease condition.
  • kits for quantitatively determining hepcidin levels which comprises obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to a peptide selected from the group consisting of SEQ ID NO:3, SEQ ID NO:4, and/or SEQ ID NO:5, and quantifying the hepcidin level in the sample; wherein the non-physiological level of hepcidin is indicative of the disease condition.
  • the present invention also relates to methods, assays and kits for quantitatively determining prohepcidin or mature hepcidin levels, which comprises obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to one or more epitopes of human hepcidin precursor amino acid sequence between and including amino acids 74-81 (i.e., SEQ ID NO:3), and quantifying the hepcidin level in the sample; wherein the non- physiological level of hepcidin is indicative of the disease condition.
  • the present invention also relates to methods, assays and kits for quantitatively determining hepcidin levels, which comprises obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to one or more epitopes of human hepcidin precursor amino acid sequence between and including amino acids 60-84 (i.e., SEQ ID NO:2, representing the predominant form of mature human hepcidin), and quantifying the hepcidin level in the sample; wherein the non- physiological level of hepcidin is indicative of the disease condition.
  • the present invention concerns hepcidin regulation of iron uptake by mammalian cells and the use of hepcidin and/or hepcidin specific antibodies in the diagnosis of diseases involving disturbances of iron metabolism.
  • the diagnostic detection kits of the present invention can be particularly useful in screening the overall population of either humans or animals and identifying those subjects who have these diseases.
  • one aspect of the invention relates to a method for diagnosing a disease condition characterized by non-physiological levels of hepcidin, comprising obtaining a tissue or fluid sample from a subject; contacting the sample with an antibody or fragment thereof that specifically binds to a polypeptide from the mid-portion (amino acids 20 to 50, SEQ. ID. NO.
  • hepcidin amino acids 60 to 84, SEQ. ID. NO. 2, such as a region of the human hepcidin protein comprising amino acids 74-81 (His Arg Ser Lys Cys Gly Met Cys [SEQ ID NO:31])
  • hepcidin level using an assay based on binding of the antibody and the polypeptide; wherein the non-physiological level of hepcidin is indicative of the disease condition.
  • sensitive diagnostic methods and kits were established enabling the detection of pro-hepcidin in human plasma.
  • hepcidin e.g., HEPCIDIN-25, HEPCIDIN-22, and/or HEPCIDIN-20; see Figure 8
  • the invention opens a broad range of Therapeutic perspectives, where a hepcidin antibody and diagnostic methods (e.g., HEPCIDIN-25, HEPCIDIN-22, and/or HEPCIDIN-20; see FIG. 8 ) and kits can be used for the determination of hepcidin as a parameter for the progress of the diseases mentioned above during and after therapy.
  • the present application further provides important data that hepcidine is expressed in ⁇ -cells of the islets of Langerhans in the human pancreas and that it co-localizes with insulin.
  • This application also discloses that the production of hepcidin is regulated by glucose and iron, therapeutic applications of which are discussed further herein.
  • the invention will be described in terms of: (a) generating a hepcidin protein or fragments thereof (including mature 20-25 amino acid long hepcidin protein); (b) generating antibodies that specifically bind a hepcidin protein (including antibodies which bind only pro-hepcidin and antibodies which bind the mature 20-25 amino acid long hepcidin); (c) diagnostic assays and kits for diagnosing subtyping or monitoring the diseases described herein; (d) methods for over expressing and down regulating hepcidin; and (e) therapeutic treatment of the diseases described herein.
  • hepcidin protein is defined as any mammalian hepcidin polypeptide sharing about 80 percent amino acid sequence identity with the predicted amino acid sequence published by Pigeon and co-workers ((2001) J. Biol. Chem. 276, 7811-7819 ). The term refers to both pro-hepcidin and the 20-25 amino acid mature human hepcidine, or any fragment thereof (see FIG. 8 ).
  • the hepcidin proteins provided herein also include proteins characterized by amino acid sequences similar to those of purified hepcidin proteins but into which modification are naturally provided or deliberately engineered. For example, modifications in a hepcidin peptide or DNA sequences can be made by those skilled in the art using known techniques.
  • Modifications of interest in a hepcidin protein sequences may include the alteration, substitution, replacement, insertion or deletion of a selected amino acid residue in the coding sequence.
  • one or more of the cysteine residues may be deleted or replaced with another amino acid to alter the conformation of the molecule (e.g., as shown herein regarding SEQ ID NOS:3, 4 and 5).
  • Techniques for such alteration, substitution, replacement, insertion or deletion are well known to those skilled in the art (see, e.g., U.S. Pat. No. 4,518,584 ).
  • such alteration, substitution, replacement, insertion or deletion retains the desired activity of the protein.
  • Regions of a hepcidin protein that are important for the protein function can be determined by various methods known in the art including the alanine-scanning method which involved systematic substitution of single or strings of amino acids with-alanine, followed by testing the resulting alanine-containing variant for biological activity. This type of analysis determines the importance of the substituted amino acid(s) in biological activity.
  • Production of a hepcidin protein may be accomplished by isolating a hepcidin protein from the tissue, blood or body fluids of humans or animals suffering from hemochromotosis, iron deficiency anemia, hemosiderosis, liver cirrhosis and other such diseases described herein; using standard techniques known by those of skill in the art.
  • Such techniques included in the invention also relate to methods for producing a hepcidin protein comprising growing a culture of host cells in a suitable culture medium, and purifying a hepcidin protein from the cells or the culture in which the cells are grown.
  • hepcidin protein can also be produced by chemical synthesis of the amino acid sequence of a hepcidin protein ( Pigeon et al., (2001) J. Biol. Chem. 276, 7811-7819 ), as predicted from the cloning and sequencing of a cDNA coding for a hepcidin protein.
  • This hepcidin protein sequence information may be utilized to predict the appropriate amino sequence of a fragment of a hepcidin protein to be chemically synthesized using standard peptide synthesis methods known in the art. These methods include a solid-phase method devised by R.
  • one or more reverse-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a hepcidin protein.
  • RP-HPLC reverse-phase high performance liquid chromatography
  • Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a substantially homogeneous isolated recombinant hepcidin protein.
  • a hepcidin protein thus 'purified' or 'isolated' is substantially free of other mammalian proteins and is defined in accordance with the present invention as an isolated protein.
  • sequence of a hepcidin protein may be identified using the Edman degradation method of protein sequencing. This method sequentially removes one amino acid residue at a time from the amino terminal end of a peptide for subsequent sequence identification by chromatographic procedures. See for example, the techniques described in Konigsberg and Steinman, (1977) Strategy and Methods of Sequence Analysis, in Neurath and Hill (eds.), The Proteins (3rd ed.) Vol. 3, pp. 1-178, Academic Press .
  • sequence analysis of a hepcidin protein may be accelerated by using an automated liquid phase amino acid sequenator following described techniques ( Hewick et al., (1981) J. Biol. Chem., 256:7990-7997 ; Stein and Undefriend, (1984) Analy. Chem., 136:7-23 ), thereby allowing for the analysis of picomolar quantities of a hepcidin protein.
  • the purified hepcidin protein can be used in in vitro binding assays that are well known in the art to identify molecules that bind to a hepcidin protein. These molecules include but are not limited to, for e.g., small molecules, molecules from combinatorial libraries, antibodies or other proteins.
  • the molecules identified in the binding assay are then tested for agonist or antagonist activity in in vivo tissue culture or animal models that are well known in the art. In brief, the molecules are titrated into a plurality of cell cultures or animals and then tested for either cell/animal death or prolonged survival of the animal/cells.
  • binding molecules may be complexed with toxins, e.g., ricin or cholera, or with other compounds that are toxic to cells.
  • toxins e.g., ricin or cholera
  • the toxin-binding molecule complex is then targeted to a tumor or other cell by the specificity of the binding molecule for a hepcidin protein.
  • hepcidin protein can be achieved by recombinant DNA technology.
  • appropriate hepcidin nucleotide coding sequences may be synthesized, cloned and expressed in appropriate host cells. Since the DNA sequence coding for a hepcidin protein is known ( Pigeon et al., (2001) J. Biol. Chem. 276, 7811-7819 ), DNA probes may be synthesized by standard methods known in the art to screen cDNA libraries prepared from liver tissue from human or animal subjects suffering from hemochromotosis, iron deficiency anemia, hemosiderosis, liver cirrhosis and other diseases described herein, for specific hepcidin protein cDNA's.
  • DNA probes can further be used to isolate the entire family of hepcidin protein genes from these cDNA libraries using methods that are well known to those skilled in the art. See, for example, the techniques described in Maniatis et al., (1982) Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., Chapter 7 .
  • Hybridization procedures are useful for the screening of recombinant clones by using labeled mixed synthetic oligonucleotide probes where each probe is potentially the complete complement of a specific DNA sequence in the hybridization sample that includes a heterogeneous mixture of denatured double-stranded DNA.
  • hybridization is preferably performed on either single-stranded DNA or denatured double-stranded DNA.
  • an expression library can be screened indirectly for a hepcidin protein of the invention having at least one epitope using antibodies to the protein.
  • Such antibodies can both be polyclonally or monoclonally derived and used to detect an expression product indicative of the presence of a hepcidin protein.
  • a lambda gt11 library is constructed and screened immunologically according to the method of Huynh, et al., (1985) (in DNA Cloning: A Practical Approach, D. M. Glover, ed., 1:49 ).
  • DNA sequences encoding a hepcidin protein can also be obtained by: (1) isolation of a double stranded DNA sequence from the genomic DNA, and (2) chemical manufacture of a DNA sequence to provide the necessary codons for the protein of interest.
  • PCR polymerase chain reaction
  • Methods that are well known to those skilled in the art can be used to construct expression vectors containing a hepcidin protein or fragments thereof coding sequences and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al., 1982, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., Chapter 12 .
  • a variety of host-expression vector systems may be utilized to express a hepcidin protein or fragment thereof. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing a coding sequence for a hepcidin protein or fragments thereof; yeast transformed with recombinant yeast expression vectors containing a coding sequence for a hepcidin protein or fragment thereof; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing a coding sequence for a hepcidin protein or fragment thereof; or animal cell systems infected with recombinant virus expression vectors (e.g., adenovirus, vaccinia virus) containing a coding sequence for a hepcidin protein or fragment thereof.
  • microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or co
  • the expression elements of these vectors vary in their strength and specificities.
  • any of a number of suitable transcription and translation elements may be used in the expression vector.
  • inducible promoters such as pL of bacteriophage lambda, plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used; when cloning in insect cell systems, promoters such as the baculovirus polyhedrin promoter may be used; when cloning in mammalian cell systems, promoters such as the adenovirus late promoter or the vaccinia virus 7.5K promoter may be used. Promoters produced by recombinant DNA or synthetic techniques may also be used to provide for transcription of the inserted coding sequence for a hepcidin protein or fragment thereof.
  • yeast a number of vectors containing constitutive or inducible promoters may be used.
  • Current Protocols in Molecular Biology Vol. 2, (1988) Ed. Ausubel et al., Greene Publish. Assoc. & Wiley Interscience Ch. 13 ; Grant et al., (1987) Expression and Secretion Vectors for Yeast, in Methods in Enzymology, Eds. Wu & Grossman, (1987) Acad. Press, N.Y., Vol. 153, pp. 516-544 ; Glover, (1986) DNA Cloning, Vol. II, IRL Press, Wash., D.C. Ch.
  • a hepcidin protein or fragment thereof sequence may be cloned behind either a constitutive yeast promoter such as ADH or LEU2 or an inducible promoter such as GAL ( Cloning in Yeast, Ch. 3, R. Rothstein (1986) In DNA Cloning Vol. 11, A Practical Approach, Ed. D M Glover, IRL Press, Wash., D.C .).
  • Constructs may contain the 5' and 3' non-translated regions of a cognate hepcidin protein mRNA or those corresponding to a yeast gene.
  • YEp plasmids transform at high efficiency and the plasmids are extremely stable.
  • vectors may be used which promote integration of foreign DNA sequences into the yeast chromosome.
  • a particularly good expression system that could be used to express a hepcidin protein or fragments thereof is an insect system.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • a hepcidin protein or fragment thereof coding sequence may be cloned into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • Successful insertion of the polyhedrin gene results in production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene).
  • a hepcidin protein or fragment thereof coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vivo or in vitro recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing a hepcidin protein of fragment thereof in infected hosts.
  • the vaccinia 7.5K promoter may be used. (e.g., see Mackett et al., (1982) Proc. Natl. Acad. Sci., (USA) 79:7415-7419 ; Mackett et al., (1984) J. Virol., 49:857-864 ; Panicali et al., (1982) Proc. Natl. Acad. Sci., 79: 4927-4931 ).
  • Specific initiation signals may also be required for efficient translation of the inserted hepcidin protein or fragment thereof coding sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where the entire hepcidin protein genome, including its own initiation codon and adjacent sequences, are inserted into the appropriate expression vectors, no additional translational control signals may be needed. However, in cases where only a portion of a hepcidin protein coding sequence is inserted, exogenous translational control signals, including the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of a hepcidin protein or fragment thereof coding sequence to ensure translation of the entire insert.
  • exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • the efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al., (1987) Methods in Enzymol., 153:516-544 ).
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired.
  • Expression driven by certain promoters can be elevated in the presence of certain inducers, (e.g., zinc and cadmium ions for metallothionein promoters). Therefore, expression of the genetically engineered hepcidin protein or fragment thereof may be controlled. This is important if the protein product of the cloned foreign gene is lethal to host cells.
  • modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • the host cells which contain a hepcidin protein or fragment thereof coding sequence and which express the biologically active hepcidin protein or fragment thereof gene product may be identified by at least four general approaches: (a) DNA-DNA hybridization; (b) the presence or absence of "marker" gene functions; (c) assessing the level of transcription as measured by expression of hepcidin protein mRNA transcripts in host cells; and (d) detection of hepcidin protein gene products as measured by immunoassays or by its biological activity.
  • the presence of a hepcidin protein or fragment thereof coding sequence inserted in the expression vector can be detected by DNA-DNA hybridization using probes comprising nucleotide sequences that are homologous to a hepcidin protein coding sequence or particular portions thereof substantially as described recently ( Pigeon et al., (2001) J. Biol. Chem. 276, 7811-7819 ).
  • the recombinant expression vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, resistance to methotrexate, transformation phenotype, occlusion body formation in baculovirus, etc.). For example, if a hepcidin protein or fragment thereof coding sequence is inserted within a marker gene sequence of the vector, recombinants containing a hepcidin protein or fragment thereof coding sequence can be identified by the absence of the marker gene function.
  • marker gene functions e.g., thymidine kinase activity, resistance to antibiotics, resistance to methotrexate, transformation phenotype, occlusion body formation in baculovirus, etc.
  • a marker gene can be placed in tandem with a hepcidin protein or fragment thereof coding sequence under the control of the same or different promoter used to control the expression of a hepcidin coding sequence. Expression of the marker in response to induction or selection indicates expression of a hepcidin protein coding sequence.
  • transcriptional activity for a hepcidin protein or fragment thereof coding region can be assessed by hybridization assays.
  • RNA can be isolated and analyzed by Northern blot using a probe homologous to a hepcidin protein or fragment thereof coding sequence or particular portions thereof substantially as described ( Pigeon et al., (2001) J. Biol. Chem. 276, 7811-7819 ).
  • total nucleic acids of the host cell may be extracted and assayed for hybridization to such probes.
  • the expression of a hepcidin protein or fragment thereof product can be assessed immunologically, for example by Western blots, immunoassays such as radioimmunoprecipitation, enzyme-linked immunoassays and the like.
  • the gene product should be analyzed. This can be achieved by assays based on the physical, immunological or functional properties of the product. For example, methods are described which include a process for producing a hepcidin protein in which a host cell containing a suitable expression vector that includes a hepcidin polynucleotide of the invention is cultured under conditions that allow expression of the encoded hepcidin protein.
  • a hepcidin protein can be recovered from the culture, conveniently from the culture medium, or from a lysate prepared from the host cells and further purified.
  • the protein produced by such process is a full length or mature form of the protein.
  • Isolated hepcidin protein encoded by the nucleic acid fragments herein described or by degenerate variants of the nucleic acid fragments herein described are intended nucleotide fragments that differ from a nucleic acid fragment (e.g., an ORF) by nucleotide sequence but, due to the degeneracy of the genetic code, encode an identical protein sequence.
  • Preferred nucleic acid fragments are the Orbs that encode proteins.
  • a hepcidin protein of the present invention can alternatively be purified from cells that have been altered to express a hepcidin protein.
  • a cell is altered to express a desired polypeptide or protein when the cell, through genetic manipulation, is made to produce a hepcidin protein which it normally does not produce or which the cell normally produces at a lower level.
  • One skilled in the art can readily adapt procedures for introducing and expressing either recombinant or synthetic sequences into eukaryotic or prokaryotic cells in order to generate a cell which produces a hepcidin protein of the present invention.
  • a hepcidin protein of the invention may also be expressed as a product of transgenic animals, e.g., as a component of the milk of transgenic cows, goats, pigs, or sheep which are characterized by somatic or germ cells containing a nucleotide sequence encoding a hepcidin protein.
  • a hepcidin protein (e.g., see SEQ ID NOS. 1-7) may also be produced by known conventional chemical synthesis. Methods for constructing a hepcidin protein of the present invention by synthetic means are known to those skilled in the art.
  • the synthetically-constructed hepcidin protein sequences by virtue of sharing primary, secondary or tertiary structural and/or conformational characteristics with natural hepcidin protein may possess biological properties in common therewith, including protein activity. Thus, they may be employed as biologically active or immunological substitutes for a natural, purified hepcidin protein in screening of therapeutic compounds and in immunological processes for the development of antibodies.
  • a hepcidin protein of the invention may be prepared by culturing transformed host cells under culture conditions suitable to express the recombinant protein.
  • the resulting expressed hepcidin protein may then be purified from such culture (i.e., from culture medium or cell extracts) using known purification processes, such as gel filtration and ion exchange chromatography.
  • the purification of a hepcidin protein may also include an affinity column containing agents which will bind to the protein; one or more column steps over such affinity resins as concanavalin A-agarose, heparin-toyopearl" or Cibacrom blue 3GA SepharoseTM; one or more steps involving hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether; or immunoaffinity chromatography.
  • affinity resins as concanavalin A-agarose, heparin-toyopearl" or Cibacrom blue 3GA SepharoseTM
  • hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether
  • immunoaffinity chromatography immunoaffinity chromatography
  • a hepcidin protein of the invention may also be expressed in a form that will facilitate purification.
  • it may be expressed as a fusion protein, such as those of maltose binding protein (MBP), glutathione-S-transferase (GST) or thioredoxin (TRX), or as a His tag. Kits for expression and purification of such fusion proteins are commercially available from New England BioLab (Beverly, Mass.), Pharmacia (Piscataway, N.J.) and Invitrogen, respectively.
  • a hepcidin protein can also be tagged with an epitope and subsequently purified by using a specific antibody directed to such epitope.
  • FLAG ® is commercially available from Kodak (New Haven, Conn.).
  • a hepcidin protein or fragment thereof should be immunoreactive whether it results from the expression of the entire gene sequence, a portion of the gene sequence or from two or more gene sequences which are ligated to direct the production of chimeric proteins. This reactivity may be demonstrated by standard immunological techniques, such as radioimmunoprecipitation, radioimmune competition, or immunoblots.
  • the immunization peptide was coupled to keyhole limpet hemocyanin using m-maleimidobenzoyl- N -hydroxysuccinimide ester, and mice were immunized with the peptide conjugate.
  • mice were immunized with the peptide conjugate.
  • the antibodies mHK(5), mHK(8/1), mHK(8/2), mHK(8/3), and mHK(9), each directed against hepcidin-(74-Abu-81), were used in studies, as discussed herein. These antibodies were able to identify hepcidin in Western blot, immunohistochemistry and ELISA experiments.
  • the hybridoma cells which produce the antibody mHK(9) have been deposited at the German Collection of Microorganisms and Cell Cultures (i.e. DSMZ; Deutsche Sammlung von Mikroorganismen und Zellkulturen, GmbH) in Braunschweig, Germany under the accession number DSM ACC2812.
  • DSMZ German Collection of Microorganisms and Cell Cultures
  • the depositor and address of DSM ACC2812 is Dr. med. Hasan Kulalsiz; Steubenstr. 8; 69121 Heidelberg, Germany.
  • hepcidin specific antibodies bind those epitopes and no other known sequences.
  • the epitope is identical to SEQ. ID. NO. 4.
  • SEQ. ID. NO. 4 is cysteine (i.e., SEQ ID NO:3).
  • cysteine In different embodiments, the cysteine"x" at position 5 of this sequence (HRSKXGMC (SEQ ID NO:4) is replaced with another molecule, which preferably resembles cysteine but cannot form disulfide bridges with the cysteine at the C-terminus of SEQ. ID. NO. 3.
  • the residue designated as "X” is an alpha aminobutyric acid (see SEQ ID NO:5).
  • Suitable non-limiting examples of such molecules are in general, alpha-amino acids with straight or branched backbones up to 9 carbon atoms long. These alpha amino acids may have hydroxy groups attached to carbon atoms ⁇ to ⁇ .
  • a person of the ordinary skill in the art will be able to select appropriate residues taking into consideration the following factors: structural similarity with cysteine and inability (or limited ability) to form bonds other than peptide bonds with immediately adjacent amino acids (in this case, lysine and glycine).
  • Such antibodies include but are not limited to polyclonal, monoclonal, chimeric, humanized, human, single chain, Fab fragments and an Fab expression library.
  • various host animals may be immunized by injection with a particular hepcidin protein, or a synthetic hepcidin protein or an immunogenic peptide comprising a portion of hepcidin (e.g., see SEQ ID NOS:3, 4 and/or 5), including but not limited to rabbits, mice, rats, etc.
  • adjuvants may be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • corynebacterium parvum corynebacterium parvum
  • antibodies may take the form of any type of relevant antibody fragment, antibody binding portion, specific binding member, a non-protein synthetic mimic, or any other relevant terminology known in the art which refers to an entity which at least substantially retains the binding specificity/neutralization activity. Therefore, the term "antibody” as used in any context within this specification is meant to include, but not be limited to, any specific binding member, immunoglobulin class and/or isotype (e.g., IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, IgE and IgM); and biologically relevant fragment or specific binding member thereof, including but not limited to Fab, F(ab')2, Fv, and scFv (single chain or related entity), which are capable of binding to the respective hepcidin protein or fragment, including but not limited to pro-hepcidin and any mature form of hepcidin.
  • immunoglobulin class and/or isotype e.g., IgG1, IgG
  • an "antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • a heavy chain is comprised of a heavy chain variable region (VH) and a heavy chain constant region (CHI, CH2 and CH3).
  • a light chain is comprised of a light chain variable region (Vat) and a light chain constant region (CL).
  • the variable regions of both the heavy and light chains comprise a framework (FW) and complementarily determining regions (CDR).
  • the four (4) FW regions are relatively conversed while CDR regions (CDR1, CDR2 and CDR3) represent hypervariable regions and are arranged from NH 2 terminus to the COOH terminus as follows: FW1, CDR1, FW2, CDR2, FW3, CDR3, FW4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen while, depending of the isotype, the constant region(s) may mediate the binding of the immunoglobulin to host tissues or factors. That said, also included in the working definition of "antibody” are chimeric antibodies, humanized antibodies, a recombinant antibody, human antibodies generated from a transgenic non-human animal, as well as antibodies selected from libraries using enrichment technologies available to the artisan.
  • an “antibody” is any such entity or specific binding member, which specifically binds to the respective portion of hepcidin so as to be useful in various diagnostic methods, as well as being a candidate for therapeutic applications intended to treat non-physiological concentrations of hepcidin protein within the patient, such as abnormally high concentrations of hepcidin associated with numerous disease states disclosed herein. Therefore, the term “antibody” further describes an immunoglobulin, whether natural or partly or wholly synthetically produced; any polypeptide or protein having a binding domain which is, or is substantially homologous to, an antibody binding domain.
  • antibodies can be derived from natural sources, or they may be partly or wholly synthetically produced.
  • antibodies are the immunoglobulin isotypes and their isotypic subclasses; fragments which comprise an antigen binding domain such as Fab, scFv, Fv, dAb, Fd and diabodies, as discussed without limitation, infra. It is known in the art that it is possible to manipulate monoclonal and other antibodies and use techniques of recombinant DNA technology to produce other antibodies or chimeric molecules which retain the specificity of the original antibody.
  • Such techniques may evolve introducing DNA encoding the immunoglobulin variable region, or the complementarily determining regions (CDRs), of an antibody to the constant regions, or constant regions plus framework regions, of a different immunoglobulin.
  • a hybridoma or other cell producing an antibody may be subject to genetic mutation or other changes, which may or may not alter the binding specificity of antibodies produced.
  • Antibodies can be modified in a number of ways, and the term "antibody” should be construed as covering any specific binding member or substance having a binding domain with the required specificity, as shown herein with the exemplified mouse monoclonal antibodies.
  • this term covers antibody fragments, derivatives, functional equivalents and homologues of antibodies, including any polypeptide comprising an immunoglobulin binding domain, whether natural or wholly or partially synthetic.
  • an entity may be a binding fragment encompassed within the term "antigen-binding portion" or "specific binding member” of an antibody including but not limited to (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment, which comprises a VH domain; (vi) an isolated complementarily determining region (CDR); (vii) a 'scAb', an antibody fragment containing V
  • Polyclonal antibodies may be readily generated by one of ordinary skill in the art from a variety of warm-blooded animals such as horses, cows, various fowl, rabbits, mice, or rats. Briefly, hepcidin is utilized to immunize the animal through intraperitoneal, intramuscular, intraocular, or subcutaneous injections, an adjuvant such as Freund's complete or incomplete adjuvant. Following several booster immunizations, samples of serum are collected and tested for reactivity to hepcidin. Particularly preferred polyclonal antisera will give a signal on one of these assays that is at least three times greater than background. Once the titer of the animal has reached a plateau in terms of its reactivity to hepcidin, larger quantities of antisera may be readily obtained either by weekly bleedings, or by exsanguinating the animal.
  • Monoclonal antibodies to peptides of hepcidin may be prepared by using any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Kohler and Milstein, (Nature, (1975) 256:495-497 ), the more recent human B-cell hybridoma technique ( Kosbor et al., (1983) Immunology Today, 4:72 ) and the EBV-hybridoma technique ( Cole et al., (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 ).
  • monoclonal antibodies specific to hepcidin proteins/peptides may be produced in germ-free animals utilizing recent technology ( PCT/US90/02545 ).
  • human antibodies may be used and can be obtained by using human hybridomas ( Cote at al., (1983) Proc. Natl. Acad. Sci., 80:2026-2030 ) or by transforming human B cells with EBV virus in vitro ( Cole et al., (1985) in, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, pp. 77-96 ).
  • techniques developed for the production of "chimeric antibodies” Morrison et al., (1984) Proc. Natl.
  • polyclonal or monoclonal antibodies for use in the disclosed treatment methods may be raised by known techniques.
  • Monospecific (i.e., monoclonal) murine (mouse) antibodies showing specificity to a confirmational epitope may be purified from mammalian antisera containing antibodies reactive against this region, or may be prepared as monoclonal antibodies using any techniques available to the artisan.
  • "Monospecific” or “monoclonal” antibody as used herein is defined as a single antibody species or multiple antibody species with homogenous binding characteristics.
  • hybridoma cells are produced by mixing the splenic lymphocytes with an appropriate fusion partner, preferably myeloma cells, under conditions which will allow the formation of stable hybridomas.
  • the splenic antibody producing cells and myeloma cells are fused, selected, and screened for antibody production.
  • Hybridoma cells from antibody positive wells are cloned by a_technique such as the soft agar technique of MacPherson, 1973, Soft Agar Techniques, in Tissue Culture Methods and Applications, Kruse and Paterson, Eds, Academic Press .
  • Monoclonal antibodies are produced in vivo by injecting respective hydridoma cells into pristine primed mice, collecting ascite fluid after an interval of time, and prepared by techniques well known in the art.
  • the antibodies of the present invention may also be in the form of a "chimeric antibody", a monoclonal antibody constructed from the variable regions derived from say, the murine source, and constant regions derived from the intended host source (e.g., human; for a review, see Morrison and Oi, 1989, Advances in Immunology, 44: 65-92 ).
  • the variable light and heavy genes from the rodent (e.g., mouse) antibody are cloned into a mammalian expression vector which contains an appropriate human light chain and heavy chain coding region, respectively.
  • These heavy and light "chimeric" expression vectors are cotransfected into a recipient cell line and selected and expanded by known techniques.
  • This cell line may then be subjected to known cell culture techniques, resulting in production of both the light and heavy chains of a chimeric antibody.
  • chimeric antibodies have historically been shown to have the antigen-binding capacity of the original rodent monoclonal while significantly reducing immunogenicity problems upon host administration.
  • a logical improvement to the chimeric antibody is the "humanized antibody,” which arguably reduces the chance of the patient mounting an immune response against a therapeutic antibody when compared to use of a chimeric or full murine monoclonal antibody.
  • the strategy of "humanizing" a murine Mab is based on replacing amino acid residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grafting of entire complementarily determining regions ( Jones et al., 1986, Nature 321: 522-526 ).
  • humanized forms of non-human (e.g., murine) antibodies are chimeric antibodies which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which hypervariable region residues of the recipient are replaced by hypervariable region residues from a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • the humanization techniques are well known in the art. Further, some humanization protocols are commercially available, for example, from Diversa Corp (San Diego, CA). In some instances, FV framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues which are not found in the recipient antibody or in the donor antibody, these modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human 'immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (FC), typically that of a human immunoglobulin.
  • FC immunoglobulin constant region
  • humanizing involves assembly of hypervariable regions of a non-human antibody and conserved regions of human antibodies.
  • An additional embodiment of the invention utilizes the techniques described for the construction of Fab expression libraries ( Huse et al., (1989) Science, 246:1275-1281 ) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity to hepcidin proteins/peptides.
  • Antibody fragments that contain specific binding sites for a hepcidin protein may be generated by known techniques.
  • such fragments include but are not limited to: the F(ab') 2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments.
  • Phage display technology is known in the art (e.g., see technology from Cambridge Antibody Technology (CAT)) as disclosed in U.S. Patent Nos. 5,565,332 ; 5, 733,743 ; 5,871,907 ; 5,872,215 ; 5,885,793 ; 5,962,255 ; 6,140,471 ; 6,225,447 ; 6,291650 ; 6,492,160 ; 6,521,404 ; 6,544,731 ; 6,555,313 ; 6,582,915 ; 6,593, 081 , as well as other U.S. family members, or applications which rely on priority filing GB 9206318, filed 24 May 1992 ; see also Vaughn, et al.
  • Single chain antibodies may also be designed and constructed using available recombinant DNA technology, such as a DNA amplification method (e.g., PCR), or possibly by using a respective hybridoma cDNA as a template.
  • Single-chain antibodies can be mono-or bispecific; bivalent or tetravalent.
  • a nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below.
  • recombinant human antibody represents a viable subset of "antibodies” generated by various means of recombinant DNA technology and non-human transgenics that are well known in the art. Such methodology is utilized to generate an antibody from one or the following origins: (i) a scFv or alternative antibody isolated from a combinatorial human antibody library; (ii) a partial or complete antibody generated from a respective expression vector stably or transiently transfected into a host cell, preferably a mammalian host cell; and/or (iii) an antibody isolated from a non-human transgenic animal which contains human immunoglobulin genes, or by any other known methodology which relies of the recombinant 'mixing and matching' of human immunoglobulin gene sequences to other DNA sequences in order to generate the human recombinant antibody of interest.
  • the invention provides a method of monitoring a disease condition characterized by non-physiological levels of hepcidin in a subject, comprising: determining an amount of the hepcidin protein (including both 84 amino acid long hepcidin and the 20-25 amino acid long mature hepcidin) at a first time; determining an amount of the hepcidin protein at a second, later time; whereby
  • This method may be applied to follow a course of any of the disease conditions described herein, with representative non-limiting examples being anemia of chronic disease and renal insufficiency.
  • the method of the instant aspect may be used to evaluate a treatment of any of the diseases.
  • the measurement of hepcidin conducted at the first time is prior to the treatment, and the measurement of hepcidin conducted at the second time is during or after the treatment.
  • Suitable examples of treatments include, without limitation erythropoietin therapy and/or iron substitution (e.g., oral or i.v. iron substitution).
  • a normal ranges or amounts of hepcidin are available from public sources, such as, for example, Pubmed.
  • the level of the 84 amino acid hepcidin was 92 ⁇ 17 ng/ml (mean ⁇ SE; 10 healthy persons).
  • the physiological concentration of 84-amino acid long hepcidin is in the range of 51.6-153.4 ng/mL (mean 106.2 ng/mL).
  • the normal range or level may be compared with the level of hepcidin during or after the treatment and if a significant difference is found, then the practitioner may infer the efficacy of the treatment.
  • Yet another purpose of the present invention is to provide reagents for use in diagnostic assays for the detection of a hepcidin protein (including both pro-hepcidin and the 20-25 amino acid long mature hepcidin) and hepcidin fragments from individuals suffering from hemochromotosis, iron deficiency anemia, hemosiderosis, liver cirrhosis and such other diseases described herein.
  • a hepcidin protein including both pro-hepcidin and the 20-25 amino acid long mature hepcidin
  • hepcidin fragments from individuals suffering from hemochromotosis, iron deficiency anemia, hemosiderosis, liver cirrhosis and such other diseases described herein.
  • a hepcidin protein of the present invention may be used as an antigen in immunoassays for the detection of those individuals suffering from hemochromotosis, iron deficiency anemia, hemosiderosis, liver cirrhosis and such other diseases described herein.
  • a hepcidin protein, polypeptide and/or peptide of the present invention may be used in any immunoassay system known in the art including, but not limited to: radioimmunoassays, enzyme-linked immunosorbent assay, "sandwich” assays, precipitin reactions, gel diffusion immunodiffusion assays, agglutination assays, fluorescent immunoassays, protein A immunoassays and immunoelectrophoresis assays, to name but a few U.S. Pat. No. 4,629,783 and patents cited therein also describe suitable assays.
  • monoclonal or polyclonal antibodies produced to various forms of a hepcidin protein can be used in an immunoassay on samples of blood, spinal fluid or other body fluid to diagnose subjects with hemochromotosis, iron deficiency anemia, hemosiderosis, liver cirrhosis and other diseases described herein.
  • the immunoassays are performed in vitro.
  • a sample of blood is removed from the patient by venesection and placed in contact with an anticoagulant such as EDTA, mixed, centrifuged at 600 g for 10 min and the plasma removed as is common in the art or a sample of spinal fluid is removed from the patient by lumbar puncture.
  • an anticoagulant such as EDTA
  • the antibodies described herein may be used as the basic reagents in a number of different immunoassays to determine the presence of a hepcidin protein in a sample of tissue, blood or body fluid.
  • the antibodies can be employed in any type of immunoassay, whether qualitative or quantitative. This includes both the two-site sandwich assay and the single site immunoassay of the non-competitive type, as well as in traditional competitive binding assays.
  • sandwich or double antibody assay of which a number of variations exist, all of which are intended to be encompassed by the present invention.
  • unlabeled antibody is immobilized on a solid substrate, e.g., microtiter plate wells, and the sample to be tested is brought into contact with the bound molecule.
  • a second antibody labeled with a reporter molecule capable of inducing a detectable signal, is then added and incubation is continued allowing sufficient time for binding with the antigen at a different site and the formation of a ternary complex of antibody-antigen-labeled antibody.
  • any unreacted material is washed away, and the presence of the antigen is determined by observation of a signal, which may be quantitated by comparison with a control sample containing known amounts of antigen.
  • Variations on the forward sandwich assay include the simultaneous assay, in which both sample and antibody are added simultaneously to the bound antibody, or a reverse sandwich assay in which the labeled antibody and sample to be tested are first combined, incubated and added to the unlabelled surface bound antibody.
  • the only limiting factor is that both antibodies have different binding specificities for a hepcidin protein.
  • both antibodies have different binding specificities for a hepcidin protein.
  • a primary antibody is either covalently or passively bound to a solid support.
  • the solid surface is usually glass or a polymer, the most commonly used polymers being cellulose, polyacrylamide, nylon, polystyrene, polyvinylchloride or polypropylene.
  • the solid supports may be in the form of tubes, beads, discs or microplates, or any other surfaces suitable for conducting an immunoassay.
  • the binding processes are well known in the art. Following binding, the solid phase-antibody complex is washed in preparation for the test sample. An aliquot of the body fluid containing a hepcidin protein to be tested is then added to the solid phase complex and incubated at 25°C.
  • reporter molecule a molecule which by its chemical nature, provides an analytically detectable signal which allows the detection of antigen-bound antibody. Detection must be at least relatively quantifiable, to allow determination of the amount of antigen in the sample, this may be calculated in absolute terms, or may be done in comparison with a standard (or series of standards) containing a known normal level of antigen.
  • reporter molecules in this type of assay are either enzymes or fluorophores.
  • an enzyme immunoassay an enzyme is conjugated to the second antibody, often by means of glutaraldehyde or periodate.
  • Commonly used enzymes include horseradish peroxidase, glucose oxidase, beta-galactosidase and alkaline phosphatase, among others.
  • the substrates to be used with the specific enzymes are generally chosen for the production, upon hydrolysis by the corresponding enzyme, of a detectable color change.
  • p-nitrophenyl phosphate is suitable for use with alkaline phosphatase conjugates; for peroxidase conjugates, 1,2-phenylenediamine or toluidine are commonly used.
  • fluorogenic substrates which yield a fluorescent product rather than the chromogenic substrates noted above.
  • the enzyme-labeled antibody is added to the first antibody-hepcidin protein complex and allowed to bind to the complex, and then the excess reagent is washed away. A solution containing the appropriate substrate is then added to the tertiary complex of antibody-antigen-labeled antibody.
  • the substrate reacts with the enzyme linked to the second antibody, giving a qualitative visual signal, which may be further quantitated, usually spectrophotometrically, to give an evaluation of the amount of antigen that is present in the serum sample.
  • fluorescent compounds such as fluorescein or rhodamine
  • fluorescein or rhodamine may be chemically coupled to antibodies without altering their binding capacity.
  • the fluorochrome-labeled antibody When activated by illumination with light of a particular wavelength, the fluorochrome-labeled antibody absorbs the light energy, inducing a state of excitability in the molecule, followed by emission of the light at a characteristic longer wavelength. The emission appears as a characteristic color visually detectable with a light microscope.
  • EIA enzyme immunoassay
  • the fluorescent-labelled antibody is allowed to bind to the first antibody-hepcidin protein complex. After washing the unbound reagent, the remaining ternary complex is then exposed to light of the appropriate wavelength, and the fluorescence observed indicates the presence of the antigen.
  • Immunofluorescence and EIA techniques are both very well established in the art and are particularly preferred for the present method.
  • reporter molecules such as radioisotopes, chemiluminescent or bioluminescent molecules may also be employed. It will be readily apparent to the skilled artisan how to vary the procedure to suit the required use.
  • the sample to be tested either human blood or spinal fluid containing a hepcidin protein may be used in a single site immunoassay wherein it is adhered to a solid substrate either covalently or noncovalently.
  • An unlabeled anti-hepcidin protein antibody is brought into contact with the sample bound on the solid substrate.
  • a second antibody labelled with a reporter molecule capable of inducing a detectable signal, is then added and incubation is continued allowing sufficient time for the formation of a ternary complex of antigen-antibody-labeled antibody.
  • the second antibody may be a general antibody (i.e., zenogeneic antibody to immunoglobulin, particularly anti-(IgM and IgG) linked to a reporter molecule) that is capable of binding an antibody that is specific for a hepcidin protein of interest.
  • a general antibody i.e., zenogeneic antibody to immunoglobulin, particularly anti-(IgM and IgG) linked to a reporter molecule
  • a hepcidin gene (mutated or normal) can be utilized in an assay of iron metabolism.
  • the gene is expressed, with or without any accompanying molecules, in cell lines or primary cells derived from human or animal subjects, healthy subjects, or cells from other organisms (such as rodents, insects, bacteria, amphibians, etc.). Uptake of iron by these cells is measured, for example through the use of radioactive isotopes. Further, binding of iron to a hepcidin gene product can also be measured. Such experiments assist in assessing the role of a hepcidin gene and hepcidin gene product in iron uptake, binding, and transport by and in cells.
  • the hepcidin diagnostic methods and kits can be used in genetic technological approaches, such as for over expressing or down regulating hepcidin.
  • down regulation of a normal hepcidin gene or a normal hepcidin protein is desirable in situations where iron is under accumulated in the body, for example in certain anemias (i.e., anemia by bleeding or renal anemia).
  • upregulation of a hepcidin gene or a hepcidin protein is desirable in situations where iron is over-accumulated in the body because hepcidin inhibits iron absorption.
  • diseases which may be suitable for treatment with hepcidin or fragments thereof are the diseases described elsewhere in this application. These diseases may optionally be accompanied by non-physiological hepcidin or prohepcidin levels. Suitable non-representative examples of the diseases include, but are in no way limited to,hemochromatosis, hemosiderosis, and primary and/or secondary iron overload.
  • antibodies specific to a normal or a mutant hepcidin protein can be prepared. Such antibodies can be used therapeutically in the diseases described herein. For example, the antibodies can be used therapeutically to block or inactivate the action of a hepcidin protein that leads to a decrease of body iron.
  • a hepcidin gene in a normal or in a mutant form, can be down regulated through the use of antisense oligonucleotides directed against the gene or its transcripts.
  • a similar strategy can be utilized as discussed above in connection with antibodies.
  • the antisense oligonucleotides herein described may be synthesized by any of the known chemical oligonucleotide synthesis methods. Such methods are generally described, for example, in Winnacker Chirurg (1992) 63:145 .
  • Antisense oligonucleotides are most advantageously prepared by utilizing any of the commercially available, automated nucleic acid synthesizers.
  • One such device, the Applied Biosystems 380B DNA Synthesizer utilizes beta-cyanoethyl phosphoramidite chemistry.
  • oligonucleotide synthesis of DNA complementary to a hepcidin gene is known, the mRNA transcript of the cDNA sequence is also known.
  • antisense oligonucleotides hybridizable with any portion of such transcripts may be prepared by oligonucleotide synthesis methods known to those skilled in the art. While any length oligonucleotide may be utilized in the practice of the invention, sequences shorter than 12 bases may be less specific in hybridizing to the target mRNA, may be more easily destroyed by enzymatic digestion, and may be destabilized by enzymatic digestion. Hence, oligonucleotides having 12 or more nucleotides are preferred.
  • oligomers of 12-40 nucleotides are preferred, more preferably 15-30 nucleotides, most preferably 18-26 nucleotides. Sequences of 18-24 nucleotides are most particularly preferred.
  • the antisense therapy may be accomplished by siRNA or shRNA treatment.
  • SiRNAs are typically short (19-29 nucleotides), double-stranded RNA molecules that cause sequence-specific degradation of complementary target mRNA known as RNA interference (RNAi) ( Bass, Nature 411:428 (2001 )).
  • RNAi RNA interference
  • the siRNA molecules comprise a double-stranded structure comprising a sense strand and an antisense strand
  • the antisense strand comprises a nucleotide sequence that is complementary to at least a portion of a desired nucleic acid sequence (i.e., hepcidin mRNA)
  • the sense strand comprises a nucleotide sequence that is complementary to at least a portion of the nucleotide sequence of said antisense region
  • the sense strand and the antisense strand each comprise about 19-29 nucleotides.
  • nucleic acid sequence can be targeted by the siRNA molecules. of the present invention.
  • Nucleic acid sequences encoding hepcidin are publicly available from Genbank.
  • the target is a human hepcidin mRNA.
  • the siRNA molecules targeted to desired sequence can be designed based on criteria well known in the art (e.g., Elbashir et al., EMBO J. 20:6877 (2001 )).
  • the target segment of the target mRNA preferably should begin with AA (most preferred), TA, GA, or CA; the GC ratio of the siRNA molecule preferably should be 45-55%; the siRNA molecule preferably should not contain three of the same nucleotides in a row; the siRNA molecule preferably should not contain seven mixed G/Cs in a row; the siRNA molecule preferably should comprises two nucleotide overhangs (preferably TT) at each 3' terminus; the target segment preferably should be in the ORF region of the target mRNA and preferably should be at least 75 bp after the initiation ATG and at least 75 bp before the stop codon; and the target segment preferably should not contain more than 16-17 contiguous base pairs of homology to other coding sequences.
  • siRNA molecules targeted to desired sequences can be designed by one of skill in the art using the aforementioned criteria or other known criteria (e.g., Gilmore et al., J. Drug Targeting 12:315 (2004 ); Reynolds et al., Nature Biotechnol. 22:326 (2004 ); Ui-Tei et al., Nucleic Acids Res. 32:936 (2004 )).
  • siRNA molecules Such criteria are available in various web-based program formats useful for designing and optimizing siRNA molecules (e.g., Sidesign Center at Dharmacon; BLOCK-IT RNAi Designer at Invitrogen; siRNA Selector at WISTAR Insitute; siRNA selection program at Whitehead Institute; siRNA Design at Integrated DNA Technologies; siRNA Target Finder at Ambion; AND siRNA Target Finder at Genscript).
  • a person of skill in the art may just find suitable siRNA sequences by entering the desired template sequence (in this case, the mRNA sequence for hepcidin, including, without limitation, human hepcidin) into one or more of the software programs listed above.
  • SiRNA molecules targeted to desired sequences can be produced in vitro by annealing two complementary single-stranded RNA molecules together (one of which matches at least a portion of a desired nucleic acid sequence) (e.g., U.S. Pat. no. 6,506,559 ) or through the use of a short hairpin RNA (shRNA) molecule which folds back on itself to produce the requisite double-stranded portion ( Yu et al., Proc. Natl. Acad. Sci. USA 99:6047 (2002 )).
  • shRNA short hairpin RNA
  • RNA molecules can be chemically synthesized (e.g., Elbashir et al., Nature _411:494 (2001 )) or produced by in vitro transcription using DNA templates (e.g., Yu et al., Proc. Natl. Acad. Sci. Usa 99:6047 (2002 )).
  • chemical modifications can be introduced into the siRNA molecules to improve biological stability.
  • modifications include phosphorothioate linkages, fluorine-derivatized nucleotides, deoxynucleotide overhangs, 2'-O-methylation, 2'-O-allylation, and locked nucleic acid (LNA) substitutions ( Dorset and Tuschl, Nat. Rev. Drug Discov. 3:318 (2004 ); Gilmore et al., J. Drug Targeting 12:315 (2004 )).
  • SiRNA molecules targeted to desired target sequences can be introduced into cells to inhibit expression.
  • the siRNA molecules are introduced into a cell expressing hepcidin (for example, a pancreatic cell or a kidney cell).
  • the synthesis of oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
  • small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 mmol scale protocol with a 2.5 min coupling step for 2'-O-methylated nucleotides and a 45 second coupling step for 2'-deoxy nucleotides or 2'-deoxy-2'-fluoro nucleotides.
  • Syntheses at the 0.2 mmol scale can be performed on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle.
  • Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by colorimetric quantitation of the trityl fractions, are typically 97.5-99%.
  • synthesizer include the following: detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methyl imidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); and oxidation solution is 16.9 mM I.sub.2, 49 mM pyridine, 9% water in THF (PerSeptive Biosystems, Inc.). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-dioxide, 0.05 M in acetonitrile) is used.
  • Deprotection of the DNA-based oligonucleotides is performed as follows: the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aqueous methylamine (1 mL) at 65 °C for 10 minutes. After cooling to -20 °C, the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H 2 O/3:1:1, vortexed and the supernatant is then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, are dried to a white power.
  • siRNA molecules can be introduced into cells in vivo by direct delivery into specific organs such as the kidneys or the pancreas, or systemic delivery into the blood stream or nasal passage using naked siRNA molecules or siRNA molecules encapsulated in biodegradable polymer microspheres ( Gilmore et al., J. Drug Targeting 12:315 (2004 )).
  • siRNA molecules targeted to specific mRNA sequences can be introduced into cells in vivo by endogenous production from an expression vector(s) encoding the sense and antisense siRNA sequences.
  • an expression vector comprising at least one DNA sequence encoding a siRNA molecule corresponding to at least a portion of hepcidin mRNA nucleic acid sequence capable of inhibiting expression of a specific mRNA in a cell operably linked to a genetic control element capable of directing expression of the siRNA molecule in a cell is described.
  • Expression vectors can be transfected into cells using any of the methods described above.
  • Genetic control elements include a transcriptional promoter, and may also include transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription.
  • Suitable eukaryotic promoters include constitutive RNA polymerase II promoters (e.g., cytomegalovirus (CMV) promoter, the SV40 early promoter region, the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (RSV), the herpes thymidine kinase (TK) promoter, and the chicken beta-actin promoter), cardiac-tissue-specific RNA polymerase II promoters (e.g., the ventricular myosin light chain 2 (MLC-2V) promoter, and the sodium-calcium exchanger gene H1 promoter (NCX1H1)), and RNA polymerase III promoters (e.g., U6, H1, 7SK AND 7SL).
  • CMV cytomegalovirus
  • the sense and antisense strands of siRNA molecules are encoded by different expression vectors (i.e., cotransfected) (e.g., Yu et al., Proc. Natl. Acad. Sci. USA 99:6047 (2002 ).
  • the sense and antisense strands of siRNA molecules are encoded by the same expression vector.
  • the sense and antisense strands can be expressed separately from a single expression vector, using either convergent or divergent transcription (e.g., Wang et al., Proc. Natl. Acad. Sci. USA 100:5103 (2003 ); Tran et al., BMC Biotechnol. 3:21 (2003 )).
  • the sense and antisense strands can be expressed together from a single expression vector in the form of a single hairpin RNA molecule, either as a short hairpin RNA (shRNA) molecule (e.g., Arts et al., Genome Res. 13:2325 (2003 )) or a long hairpin RNA molecule (e.g., Paddison et al., Proc. Natl. Acad. Sci. USA 99:1443 (2002 )).
  • shRNA short hairpin RNA
  • a long hairpin RNA molecule e.g., Paddison et al., Proc. Natl. Acad. Sci. USA 99:1443 (2002 )
  • viral expression vectors are preferred, particularly those that efficiently transduce human cells (e.g., alphaviral, lentiviral, retroviral, adenoviral, adeno-associated viral (AAV)) ( Williams and Koch, Annu. Rev. Physiol. 66:49 (2004 ); Del Monte and Hajjar, J. Physiol. 546.1:49 (2003 ).
  • alphaviral, lentiviral, retroviral, adenoviral, adeno-associated viral (AAV) Williams and Koch, Annu. Rev. Physiol. 66:49 (2004 ); Del Monte and Hajjar, J. Physiol. 546.1:49 (2003 ).
  • Desired gene products include, for example, desired mRNA and desired polypeptide, and both can be measured using methods well-known to those skilled in the art.
  • desired mRNA can be directly detected and quantified using, e.g., Northern hybridization, in situ hybridization, dot and slot blots, or oligonucleotide arrays, or can be amplified before detection and quantitation using, e.g., polymerase chain reaction (PCR), Reverse-Transcription-PCR (RT-PCR), PCR-Enzyme-Linked Immunosorbent Assay (PCR-ELISA), or ligase chain reaction (LCR).
  • PCR polymerase chain reaction
  • RT-PCR Reverse-Transcription-PCR
  • PCR-ELISA PCR-Enzyme-Linked Immunosorbent Assay
  • LCR ligase chain reaction
  • Desired polypeptide can be detected and quantified using various well-known immunological assays, such as, e.g., enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, and Western blotting.
  • immunological assays such as, e.g., enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, and Western blotting.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • immunoprecipitation immunofluorescence
  • Western blotting Western blotting.
  • Anti-hepcidin antibodies may be produced, for example, by the methods described in the instant application.
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising any one of the antibodies of the present invention for use in a method for treating a disease condition selected from the group consisting of primary or secondary iron deficiency, such as iron deficiency anemia, anemia of chronic disease, inflammations and infections, including sepsis; anemia in Crohn's disease, anemia in ulcerative colitis, and any combination thereof.
  • a disease condition selected from the group consisting of primary or secondary iron deficiency, such as iron deficiency anemia, anemia of chronic disease, inflammations and infections, including sepsis; anemia in Crohn's disease, anemia in ulcerative colitis, and any combination thereof.
  • a preferred embodiment of the pharmaceutical composition for use in a method of said treatment is the antibody mHk(9) deposited under accession number DSM ACC2812.
  • Methods of treating various diseases disclosed herein which comprises administering to a human or animal patient or subject showing non-physiological hepcidin or prohepcidin levels and/or activity a therapeutically effective amount of a molecule or substance as to modulate or promote either an increase or decrease in iron levels within the patient or subject so as to bring the level of pro-hepcidin or hepcidin to within normal physiological levels are described.
  • patients with a primary or seconday iron overload may be administered a therapeutically effective amount of hepcidin, hepcidin-fragments, hepcidin-derivatives, and/or prohepcidin, so as to effectively lower iron levels to physiological levels.
  • a patient showing decreased iron levels may be administered a molecule or substance which lowers pro-hepcidin and/or hepcidin levels and/or activity so as to effect iron retention within the patient.
  • any such therapy may be combined with administration of an iron source so as to better promote a rise in the patient's iron level to physiologically normal levels.
  • One embodiment of this portion of the invention indicates, but in no way limits, the use of these methods to treat various types of hemochromatosis and hemosiderosis. Such methodology becomes even more appropriate in view of the link between glucose and iron levels disclosed herein.
  • modulating pro-hepcidin and/or hepcidin ranges from initiating to shutting down, and within that range includes enhancing significantly or slightly to inhibiting significantly or slightly, so as to effect the level of iron within the patient.
  • inhibiting includes a down regulation which may reduce or eliminate the function of mature hepcidin. For example, a given patient's condition (iron overload) may require an initiation of hepcidin-like activity to effectively modulate iron levels in the patient to physiological levels; whereas another patients condition (iron deficiency) may require an inhibition of hepcidin-like activity so that patient may also modulate iron levels to acceptable, physiological levels.
  • subject or “patient” may be used interchangeably and shall mean any animal belonging to phylum Chordata, including, without limitation, humans.
  • treating or “treatment” of a disease refers to executing a protocol, which may include administering one or more molecules, substances, drugs, etc, to a patient or subject in an effort to alleviate signs or symptoms of the disease, such as an iron overload or iron deficiency. Alleviation can occur prior to signs or symptoms of the disease appearing, as well as after their appearance, as per results of various assays disclosed herein. Thus, “treating” or “treatment” includes “preventing” or "prevention” of disease.
  • treating does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols which have only a marginal effect on the subject(such as at least a positive movement away from non-physiological iron levels towards physiological iron levels within the patient).
  • pancreatic hepcidin in linking glucose and iron metabolisms, thus providing insight into the phenomenology of glucose/iron metabolic disorders such as in hereditary hemochromatosis, an iron overload disease with abnormal glucose homeostasis and decreased insulin secretion.
  • pancreas is one of the main organs affected by hemochromatosis
  • the expression of iron-regulatory peptide hepcidin in the pancreas opens up new vistas in the physiology of iron/glucose regulation and phenomenology of iron storing diseases at the pancreatic ß-cell level.
  • hepcidin is expressed in mammalian (e.g., rat and human) pancreatic ⁇ -cells and is co-localized with insulin in secretory granules.
  • pancreatic hepcidin is regulated by iron and glucose. More specifically, hepcidin expression was down-regulated when concentrations of a source of iron (Iron Nitrilotriacetate, or FENTA), of 65 ⁇ m or higher were used, even if the concentrations of FENTA were not cytotoxic as shown in a neutral red assay.
  • FENTA Iron Nitrilotriacetate
  • pancreatic hepcidin is induced by glucose, both in vivo and in vitro. In fact, 30 min and 60 min after oral glucose application the serum hepcidin levels increased significantly to 137 and 158%. Accordingly, a method is disclosed of increasing hepcidin concentration in a subject in need thereof, the method comprising administering the subject a therapeutic agent which is either glucose itself or which stimulates an increase in concentration of glucose in blood. Such agent may cause a release of glucose from body storages (e.g., increase production of glucose from glycogen, or the agent may metabolize into glucose. Suitable non-limiting examples of the latter therapeutic agents include carbohydrates such as sucrose or fructose. Suitable non-limiting examples of the agents which increase production of glucose from glycogen include glucagon and epinephrin.
  • Hepcidin can be used in the therapy of the disorders described herein, by treating subjects with hepcidin, and agonists or antagonists of hepcidin.
  • iron uptake in cells can be modulated by varying the concentration of hepcidin.
  • hepcidin, and agonists or antagonists of hepcidin may be useful in the treatment of conditions where there is a disturbance in iron metabolism.
  • such substances may be useful in the treatment of conditions such as haemochromatosis, neurodegenerative diseases, ischemic tissue damage, including ischemic stroke or trauma, heart disease, and tumors, in particular, skin cancer, sideroblastic anemia, thalassemia; hematologic diseases, such as leukemia, polyglobulie, macrocytic, microcytic or normocytic anemia, anemia with reticulocytosis, hemolytic anemia; disturbances of the reticuloendothelial system due to infections and diseases; inflammations and infections, including sepsis; immunologic diseases and tumors, such as carcinoma, sarcoma, lymphoma, that result in non-physiologic hepcidin concentrations; neurodegenerative diseases, such as Alzheimer's disease and Wilson's disease, renal anemia, anemia of chronic diseases, anemia in Crohns disease, anemia in ulcerative colitis, sprue, cholangitis, primary or secondary sclerosing cholangitis,
  • Methods of modulating iron metabolism using hepcidin are also described.
  • a method for treating conditions involving disturbances in iron metabolism comprising administering an iron-modulating amount of hepcidin, or a stimulant, agonist or antagonist of hepcidin is described.
  • Conditions involving disturbances in iron metabolism which may be treated using the described method include by way of example haemochromatosis, neurodegenerative diseases, ischemic tissue damage, including ischemic stroke or trauma, heart disease, and tumors, in particular skin cancer and such other diseases described herein.
  • a substance which is an agonist or antagonist of hepcidin may be identified by determining the effect of the substance on the binding activity of hepcidin to ferroportin or the effect of the substance on the expression of hepcidin in cells capable of expressing hepcidin including cells genetically engineered to express hepcidin on their surface.
  • a method is described of identifying agonists or antagonists of hepcidin comprising reacting a substance suspected of being an agonist or antagonists of hepcidin with hepcidin and ferroportin under conditions such that hepcidin is capable of binding to ferroportin; measuring the amount of hepcidin bound to ferroportin; and determining the effect of the substance by comparing the amount of hepcidin bound to ferroportin with an amount determined for a control.
  • a method is also described of identifying agonists or antagonists of hepcidin comprising reacting a substance suspected of being an agonist or antagonist of hepcidin with a cell which produces hepcidin, measuring the amount of hepcidin expressed by the cell, and determining the effect of the substance by comparing the amount of expression of hepcidin with an amount determined for a control.
  • a method for identifying an agonist or antagonist of hepcidin-mediated iron uptake comprising: incubating a cell expressing hepcidin on its surface and a substance suspected of being an agonist or antagonist of hepcidin in the presence of iron and in the absence of transferrin, measuring the amount of iron uptake into the cell, and identifying an agonist or antagonist of hepcidin-mediated iron uptake by comparing the amount of iron uptake in the cell with the amount of iron uptake in a cell from a control incubation in the absence of the substance.
  • Hepcidin peptides are provided for therapeutic use in subjects having symptoms of a primary iron overload disease or syndrome, such as hemochromatosis, or other iron overload condition caused by secondary causes, such as repeated transfusions.
  • a hepcidin peptide can be full-length hepcidin or some fragment of hepcidin.
  • a hepcidin peptide comprises the amino acid residues 28 to 47 (SEQ ID NO: 8) or 60 to 84 of a 84 amino acid long hepcidin (SEQ ID NO: 2).
  • SEQ ID NO: 8 amino acid residues 28 to 47
  • SEQ ID NO: 2 amino acid residues 28 to 47
  • SEQ ID NO: 2 The predicted amino acid sequence and genomic and cDNA sequences of hepcidin where provided in ( Krause et al., (2000) FEBS Lett.
  • a hepcidin protein or fragment thereof may be administered with beta-2-microglobulin, such as in the form of a complex. In some instances, a hepcidin protein greater than about 20 amino acids is administered in a complex with beta-2-microglobulin.
  • agonists or ancagoniscs of a hepcidin protein or a transferrin receptor are provided.
  • Agonists of a hepcidin polypeptide, and/or antagonists of a transferrin receptor are useful for example, in the treatment of primary or secondary iron overload diseases or syndromes, while antagonists of a hepcidin polypeptide, or agonists of the transferrin receptor are useful, for example, in the treatment of iron deficiency conditions, such as anemias.
  • mutant hepcidin proteins/peptides are provided which function as antagonists of the wild-type hepcidin protein.
  • Antagonists or agonists can also be antibodies, directed against a transferrin receptor, or the mid-portion (amino acids 20 to 50) or C-terminal region (amino acids 60 to 84) of a hepcidin protein.
  • hepcidin polypeptides can serve as antagonists of a transferrin receptor
  • peptidomimetics can be designed using techniques well known in the art as antagonists or agonists of a hepcidin protein and/or a transferrin receptor.
  • the antibodies of the instant invention may be used to inactivate hepcidin.
  • EcR293 cells were stably transfected with a plasmid containing an ecdysone-regulated Ferroportin-GFP (Fpn-GFP) construct ( Nemeth et al. Science 306:2090-2093, (2004 )).
  • Fpn-GFP ecdysone-regulated Ferroportin-GFP
  • Fpn-GFP was induced by ponasterone treatment for 24 h.
  • Cells were then incubated with 0.1 to 0.5 ⁇ M purified human hepcidin (from urine) for 6 h and imaged by epi-fluorescence microscopy.
  • Addition of hepcidin to Fpn-GFP expressing cells changed the distribution of Fpn-GFP from the cell surface to punctuate intracellular vesicles.
  • hepcidin was absent from the medium, there was no internalization of Fpn-GFP.
  • hepcidin Concentrations of hepcidin as low as 0.1 ⁇ M induced Fpn internalization within 1 h. Hepcidin was chemically synthesized and the same experiments were performed. Chemically synthesized hepcidin was as efficient in inducing Fpn-GFP internalization as hepcidin purified from urine.
  • Ligands for ferroportin can be screened using the techniques described herein for the ability to bind to ferroportin. Additionally, competition for hepcidin binding to ferroportin can be done using techniques well known in the art. Ligands, or more generally, binding partners for a hepcidin protein can be screened, for example, for the ability to inhibit the complexing of a hepcidin polypeptide to beta-2-microglobulin, using techniques described herein.
  • Agonists or antagonists of ferroportin are similarly utilized to increase or decrease the amount of iron transported into a cell, such as into a patient's hepatocytes or lymphocytes.
  • the efficacy of a drug, therapeutic agent, agonist, or antagonist can be identified in a screening program in which modulation is monitored in in vitro cell systems.
  • Host cell systems that express various mutant hepcidin proteins/peptides and are suited for use as primary screening systems.
  • Candidate drugs can be evaluated by incubation with these cells and measuring cellular functions dependent on a hepcidin gene or by measuring proper hepcidin protein folding or processing.
  • Such assays might also entail measuring receptor-like activity, iron transport and metabolism, gene transcription or other upstream or downstream biological function as dictated by studies of hepcidin gene function.
  • cell-free systems can be utilized.
  • Purified hepcidin protein can be reconstituted into artificial membranes or vesicles and drugs screened in a cell-free system.
  • Such systems are often more convenient and are inherently more amenable to high throughput types of screening and automation.
  • Criteria for the determination of the purity of a hepcidin protein include those standard to the field of protein chemistry. These include N-terminal amino acid determination, one and two-dimensional polyacrylamide gel electrophoresis, and silver staining.
  • the purified protein is useful for use in studies related to the determination of secondary and tertiary structure, as aid in drug design, and for in vitro study of the biological function of the molecule.
  • Drugs can be designed to modulate a hepcidin gene and a hepcidin protein activity from knowledge of the structure and function correlations of a known hepcidin protein.
  • rational drug design by use of X-ray crystallography, computer-aided molecular modeling (CAMM), quantitative or qualitative structure-activity relationship (QSAR), and similar technologies can further focus drug discovery efforts.
  • Rational design allows prediction of protein or synthetic structures that can interact with and modify a hepcidin protein activity. Such structures may be synthesized chemically or expressed in biological systems. This approach has been reviewed in Capsey et al., Genetically Engineered Human Therapeutic Drugs, Stockton Press, New York (1988 ). Further, combinatorial libraries can be designed, synthesized and used in screening programs.
  • suitable carriers, excipients, and other agents may be incorporated into the formulations to provide improved transfer, delivery, tolerance, and the like.
  • formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, (15th Edition, Mack Publishing Company, Easton, Pa. (1975 )), particularly Chapter 87, by Blaug, Seymour, therein.
  • These formulations include for example, powders, pastes, ointments, jelly, waxes, oils, lipids, anhydrous absorption bases, oil-in-water or water-in-oil emulsions, emulsions carbowax (polyethylene glycols of a variety of molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax.
  • any of the foregoing formulations may be appropriate in treatments and therapies in accordance with the present invention, provided that the active agent in the formulation is not inactivated by the formulation and the formulation is physiologically compatible.
  • Solid phase peptide synthesis developed by R. B. Merrifield, was a major breakthrough allowing for the chemical synthesis of peptides and small proteins and is well known in the art.
  • an insoluble polymer support (resin) is used to anchor the peptide chain as each additional alpha-amino acid is attached.
  • This polymer support is constructed of 20-50 ⁇ m diameter particles which are chemically inert to the reagents and solvents used in solid phase peptide synthesis. These particles swell extensively in solvents, which makes the linker arms more accessible
  • Organic linkers attached to the polymer support activate the resin sites and strengthen the bond between the (-amino acid and the polymer support. Chloromethyl linkers, which were developed first, have been found to be unsatisfactory for longer peptides due to a decrease in step yields.
  • the PAM (phenylacetamidomethyl) resin because of the electron withdrawing power of the acid amide group on the phenylene ring, provides a much more stable bond than the classical resin.
  • Another alternative resin for peptides under typical peptide synthesis conditions is the Wang resin. This resin is generally used with the FMOC labile protecting group (i.e., Fluorenylmethyloxycarbonyl).
  • FMOC is a base labile protecting group which is easily removed by concentrated solutions of amines (usually 20-55% piperidine in N-methylpyrrolidone).
  • an acid labile (or base stable) resin such as an ether resin, is desired.
  • the stable blocking group protects the reactive functional group of an amino acid and prevents formation of complicated secondary chains. This blocking group must remain attached throughout the synthesis and may be removed after completion of synthesis. When choosing a stable blocking group, the labile protecting group and the cleavage procedure to be used should be considered.
  • the stable blocking groups are removed and the peptide is cleaved from the resin to produce a "free" peptide.
  • the stable blocking groups and organic linkers are labile to strong acids such as TFA.
  • the resin is washed away and the peptide is extracted with ether to remove unwanted materials such as the scavengers used in the cleavage reaction.
  • the peptide is then frozen and lyophilized to produce the solid peptide. This is then characterized by HPLC and MALDI before being used by the investigator. It may also be necessary to purify the peptide by HPLC if higher purity is required by the investigator.
  • peptides hepcidin-(28-47) and hepcidin-(70-84) were coupled to keyhole limpet hemocyanin using m-maleimidobenzoyl-N-hydroxysuccinimide ester, and two SPF rabbits (Charles River Iffa Credo) were immunized with each peptide conjugate (Eurogentec, Seraing, Belgium).
  • hepcidin 28-47 PQQ TGQ LAE LQP QDR AGA RA SEQ. (SEQ ID NO: 8)
  • hepcidin 70-84 CGC CHR SKC GMC CKT (SEQ ID NO: 9)
  • the peptide epitopes used for the generation of the antisera displayed no homology to any hitherto reported protein as confirmed by the BLAST P2 search.
  • TfR2 mouse TfR2-alpha
  • TfR2 cytoplasmic N-terminus of mouse TfR2-alpha
  • the antibody was generated in rabbits and affinity purified.
  • RNA isolation was performed using Qiagen RNA easy kit including DNA digestion.
  • Reverse transcription (RT)-PCR analysis was performed as described previously ( Kulaksiz et al., (2002) Proc. Natl. Acad. Sci. USA 99, 6796-6801 ; Kulaksiz et al., (2002) Am. J. Pathol. 161, 655-664 ) using the following primers and specifications given in 5-3' orientation: human hepcidin (GenBank database accession no. NM0211175), 5'-CTG CAA CCC CAG GAC AGA G-3'(SEQ ID NO:10) and 5, GGA ATA AAT.
  • Human TfR2 (#AF067864), 5'-GAT TCA GGG TCA GGG AGG TG-3' (SEQ ID NO:12) and 5'-(GAA GGG GCT GTG ATT GAA GG-3'(SEQ ID NO:13); corresponding to nucleotide positions 2496-2515 and 2694-2675.
  • the human hepatoma HepG2 cells were obtained from the German Collection of Microorganisms and Cell Culture (Braunschweig, Germany) and grown at 37°C. in 5% CO2 in RPMI 1640 media (Gibco, Düsseldorf, Germany) supplemented with 10% (vol/vol) heat-inactivated FBS, penicillin (100 units/ml), and streptomycin (100 mg/ml). Cells were analyzed by RT PCR using the primer specifications mentioned above. For immunofluorescence microscopy, HepG2 cells were grown on glass slides fixed for 4 mm in methanol, and permeabilized with 0.5% Triton X-100 in PBS.
  • tissues and cells were homogenized in Tris-HCl buffer containing 100 mM NaCl, 50 mM Tris-HCl, pH 7.4, 10% glycerol, 1% Triton X-100, 2 mg/ml leupeptin, 2 mg/ml pepstatin, and 1 mM phenylmethylsulfonyl fluoride, and centrifuged at 100,000 g for 30 mm at 4°C.
  • TfR2 immunoblots were performed using 8% SDS-polyacrylamide gels. Following electrophoresis, proteins were transferred onto hydrophobic polyvinylidene fluoride-based membranes (Pall, Portsmouth, England) by semidry blotting.
  • the membranes were incubated overnight with hepcidin or TfR2 antibodies at dilutions mentioned above. After washing in Tris-buffered saline containing 10 mM Tris-HCl (pH 8.0), 150 mM NaCl, and 0.05% Tween 20, the respective immunoreactive proteins were visualized after incubation with alkaline phosphatase-conjugated goat anti-rabbit antibody (diluted 1:50,000; Sigma) using nirro blue tetrazolium and 5-bromo-4-chloro-3-indolyl phosphate as chromogens (Sigma). The immunoreaction on the Western blot was specifically blocked after preincubation of the antibodies with the corresponding peptide immunogens.
  • Tissues were fixed in 4% paraformaldehyde for 18 h at 4°C. After dehydration in graded ethanol series, the specimens were embedded in paraffin. Paraffin sections (5 m) were immunostained for hepcidin (antibodies EG(1)-HepN, Kci(2)-HepN, and EG(1) HepC, each diluted 1:2000) or TfR2 (antibody BT-TFR21-S. diluted 1:1000) by the avidin-biotin-peroxidase complex (ABC) technique and incubation sequences as previously described ( Kulaksiz et al., (2002) Proc. Natl. Acad. Sci.
  • ABS avidin-biotin-peroxidase complex
  • the sections were incubated with the respective antibodies for 24 h at 4°C, followed by incubation with biotinylated anti-rabbit IgG (Jackson Immunoresearch, West Grove, Pa., USA) for 30 min diluted 1:200. The sections were then incubated for 30 min with a preformed complex of biotin-peroxidase/streptavidin (Jackson Immunoresearch), diluted in PBS (final concentrations: biotin-peroxidase, 0.7 mg/ml; streptavidin, mg/ml). The antigen-antibody binding sites were visualized by incubation of the sections in 07 mM diaminobenzidine hydrochloride/0.0020% H 2 O 2 in 0.05 M Tris-HCl pH 7.6).
  • tissue sections from human liver were prepared with a cryotome (FrigoCut 2800E; Leica, Nussloch, Germany), air dried for 2 hours, and fixed for 10 min in cold acetone (-20°C).
  • Double-immunofluorescence labeling was performed as described previously ( Rost et al., (1999) Hepatology 29, 814-821 ) using the specific hepcidin antibodies (diluted 1:1000) and monoclonal antibody C219 (id.) raised against canalicular P-glycoproteins (Centocor, Malvern, Pa.) diluted 1:30.
  • TBST TBS with 0.05% Tween 20
  • 100 ml standard samples containing various amounts of synthetic peptides or human plasma samples 58 randomized samples from our clinical laboratory
  • N-terminally biotinylated hepcidin-(28-47) Peptide Specialty Laboratories GmbH, Heidelberg, Germany
  • biotinylated antigen-antibody complexes were detected by streptavidin-peroxidase enzyme (Dako, Hamburg, Germany) with the substrate tetramethylbenzidine (DRG); the color reaction was stopped with 0.5 N H 2 SO 4 and the extinction of the solution was read at 450/630 nm wavelength.
  • RT-FCR analysis demonstrated that hepcidin is highly expressed in human liver. Similarly, a 192-bp expected transcript was detected in HepG2 cells with an expression level comparable to human liver ( FIG. 1 ). In addition, RT-PCR analyses clearly revealed that TfR2 is highly expressed in the human liver and HepG2 cells ( FIG. 1 ). In Western blot analysis, all hepcidin antibodies [EG(1)-HepN, EG(2)-HepN, and EG(1)-HepC] coincidentally identified an immunoreactive band of about 10 kDa in extracts of human and guinea pig liver.
  • This liver peptide comigrated with an immunoreactive band recognized by a hepcidin antibodies in homogenates of HepG2 cells ( FIG. 1 ). All antibodies also identified an intensively stained band at -20 kDa in all lanes loaded with human and guinea pig liver extracts or HepG2 cell extracts. Western blot analysis of skeletal muscle extracts (control) showed neither the immunoreactive band of 10 kDa nor the strong band at 20 kDa ( FIG. 1 ).
  • hepatic lobule were heterogeneous with respect to a hepcidin immunoreactivity; within a hepatic lobule, a hepcidin immunoreactive cells were predominantly located in periportal zones, and the frequency of hepcidin-positive cells continuously decreased from the portal triads toward the central veins ( FIG. 3 ).
  • distinct intercellular differences exist between a hepcidin positive cells; while most hepatocytes were strongly positive for hepcidin, others displayed only a faint staining or were totally unreactive for hepcidin ( FIG. 3 ).
  • immunofluorescence analysis demonstrated a strong immunoreactivity for hepcidin at the basolateral membrane domain; immunoreactivity was absent from the apical membrane domain as revealed by double staining with the C219 antibody raised against canalicular P-glycoproteins ( Rost et al., (1999) Hepatology 29, 814-821 ).
  • TfR2 protein-specific antibody BT-TFR21-S detected TfR2 in human and mouse liver.
  • TfR2 was found at the basolateral membrane of hepatocytes, which revealed distinct intercellular differences concerning the intensity of immunoreactivity ( FIG. 4 ).
  • Heterogeneity was also observed within a hepatic lobule with increasing immunoreactivity from the central veins to the portal triads.
  • hepcidin peptide in HepG2 cells was verified by immunocytochemistry using the corresponding peptide-specific antibodies. All antibodies identified hepcidin by the immunofluorescence technique in HepG2 cells resulting in a granular immunoreactivity pattern ( FIG. 5 ). Coincident with the cellular localization of hepcidin, the TfR2 antibody detected TfR2 in the same cells ( FIG. 5 ).
  • a sensitive hepcidin ELISA assay with a detection limit of 0.1 ng/well of the synthetic peptide was developed with the specific N-terminal hepcidin antibody EG(2)-HepN ( FIG. 6 ).
  • ELISA analyses with this antibody revealed a concentration of hepcidin in the range from 12.1 to 471.3 ng per ml human plasma. No crossreactivity was observed when heterologous peptides were used.
  • the ELISA revealed the highest resolving power between 1 and 400 ng/ml, a range, where hepcidin concentrations in human plasma were determined.
  • RT-PCR analyses with specific primers confirmed that hepcidin is highly expressed in the human liver.
  • Three different antibodies recognizing different epitopes in a hepcidin precursor molecule concurrently identified an immunoreactive peptide of about 10 kDa by Western blot analysis in liver extracts of two species, man and guinea pig.
  • the apparent molecular mass of this immunoreactive peptide is in accordance with the molecular mass deduced for a hepcidin preprohormone from the cDNA sequence ( Pigeon et al., (2001) J. Biol. Chem. 276, 7811-7819 ).
  • a second immunoreactive band of approximately 20 kDa was detected by all hepcidin antibodies in extracts of the human and guinea pig liver but was lacking in the control tissue.
  • This immunoreactive protein may represent a hepcidin-related peptide of higher molecular mass or, because of the twofold higher molecular mass of the second peptide, it may reflect a dimeric type of hepcidin.
  • an aggregation property and a possible formation of multimers was described for hepcidin-25 but not for hepcidin-20 ( Hunter et al., (2002) J. Biol. Chem., 277:37597-37603 ).
  • hepcidin is specifically localized in hepatocytes mainly located around the portal triads; the coincident staining by different region-specific antibodies not only in the human and guinea pig liver, but also in the HepG2 cells (see below) points to hepatocytes being the source of hepcidin.
  • Hepcidin immunoreactivity decreased from the periportal zones towards the central veins. This zonation within the portal lobules may have a functional significance, since the periportal hepatocytes have first-pass access to portal veins bringing iron-rich blood from the gut.
  • distinct intercellular differences exist between hepcidin-positive cells even of the same liver acinus with respect to the density of hepcidin immunoreactivity that may reflect intercellular differences in expression or secretion of hepcidin.
  • hepcidin was concentrated at the basolateral pole of hepatocytes. No immunoreactivity was found at the apical membrane domain.
  • the discrete distribution pattern of hepcidin at the subcellular level may infer a basolaterally directed release of hepcidin into the liver sinusoids.
  • This directional secretion route is additionally substantiated by the detection of hepcidin prohormone in human plasma (see below); consequently, these findings provide further evidence that hepatocytes may regulate iron metabolism in an endocrine fashion via the secretion of the peptide hormone hepcidin.
  • RT-PCR Western blot and immunohistochemical studies at the cellular level were performed.
  • RT-PCR analyses revealed that TfR2 is highly expressed in human liver. ( Fleming et al., (2000) Proc. Natl. Acadi. Sci. USA 97, 2214-2219 ). The presence of this protein was confirmed by Western blot studies using BT-TFR21-S antibody specific to human and mouse TfR2.
  • a -105 kDa immunoreactive protein was detected in mouse liver extracts; this molecular mass of immunoreactive TfR2 is slightly larger than the expected 95 kDa ( Fleming et al., (2000) Proc. Natl. Acadi. Sci. USA 97, 2214-2219 ) and may represent some posttranslational modifications as described previously ( Kawabata et al., (2000) J. Biol. Chem. 275, 16618-16625 ). Under identical conditions, however, the TfR2-antibody identified the protein at the expected 95 kDa molecular mass and with a lower affinity the 105 kDa protein in human liver extracts. The discrepancy between the immunoblots of human and mouse liver may be due to interspecies differences.
  • TfR2 is localized to hepatocytes of human and mouse liver; coincident with the cellular distribution of hepcidin, the protein-specific antibody localized TfR2 exclusively at the basolateral membrane.
  • This type of membrane-specific association of TfR2 argues particularly for a basolateral activation of TfR2, which is involved in iron metabolism by binding diferric transferrin and mediating uptake of transferrin-bound iron from the blood into hepatocytes ( Philpott, C. C. (2002) Hepatology 35, 993-1001 ; Subramaniam et al., (2002) Cell Biochem. Biophys. 36, 235-239 ).
  • a similar lobular zonation as described for hepcidin was observed for TfR2 with decreasing immunoreactivity from the periportal zones toward the central veins.
  • RT-PCR studies using the appropriate primer specifications and combinations successfully employed in the human liver identified expression of hepcidin and TfR2 in HepG2 cells.
  • the presence of hepcidin and TfR2 in HepG2 cells was confirmed by Western blot studies that yielded immunoreactive protein bands of correct molecular weights, comigrating with the corresponding immunoreactive bands from the liver tissues.
  • the co-localization of the respective proteins in HepG2 cells was particularly substantiated by immunocytochemistry using the corresponding region and molecular domain-specific antibodies.
  • hepcidin and TfR2 are colocalized at a common cell membrane domain and reveal the same lobular distribution with a strong immunoreactivity in periportal zones, the site, where in case of mutations that abrogate expression of TfR2 ( Fleming et al., (2002) Proc. Natl. Acad. Sci. USA 99, 10653-10658 ) and hepcidin ( Nicolas et al., (2001) Proc. Natl. Acad. Sci. USA 98, 8780-8785 ) but also hepcidin ( Zhou et al., (1998) Proc. Natl. Acad. Sci.
  • hepcidin is a candidate signaling factor secreted from the liver and regulating the intestinal iron absorption.
  • hepcidin is a candidate signaling factor secreted from the liver and regulating the intestinal iron absorption.
  • there is still controversy about the existence of certain molecular forms of hepcidin in the blood Krause et al., (2000) FEBS Lett. 480, 147-150 ; Park et al., (2001) J. Biol. Chem. 276, 7806-7810 ; Hunter et al., (2002) J. Biol. Chem., M205305200 ).
  • an ELISA was developed by applying the same N-terminal antibody against hepcidin prohormone used successfully in Western blot, immunocytochemical and immunofluorescence experiments.
  • the ELISA was characterized by a high sensitivity with a detection limit of 0.1 ng/well and a powerful resolution in the range of 1 to 400 ng/ml; the range, where hepcidin concentrations were determined.
  • the concentration of pro-hepcidin was measured, ranging from 12.1 to 471.3 ng/ml, which is comparable with the concentration of known regulating peptide hormones.
  • the measured concentrations exhibit a wide range of pro-hepcidin indicating that the peptide may be subject to strong regulation.
  • Future experiments are intended to determine hepcidin concentrations in plasma of various subjects with disturbances of iron metabolism and to analyze the molecular mechanism of hepcidin regulation using the established ELISA.
  • hepcidin is translated as an 84 amino acid prepropeptide that is N-terminally processed to a 20-25 amino acid peptide (id.).
  • a strong consensus sequence for a signal sequence cleavage site is located between Gly 24 and Ser 25 that would result in a 60 residue propeptide
  • previous studies failed to isolate the larger propeptide from native sources like liver tissue and blood (Id.).
  • the abundance of propeptide convertases in the liver may inhibit the isolation of certain propeptides.
  • recent studies have shown that the human circulating form of hepcidin described by two research groups in blood ( Krause et al., (2000) FEBS Lett.
  • the present invention describes hepcidin immunoreactivity in human and guinea pig liver, where it is "localized to the basolateral membrane domain of hepatocytes. Previous studies have speculated on a possible connection between these cells and the absorptive enterocytes ( Hunter et al., (2002) J. Biol. Chem., M205305200 ; Anderson et al., (2002) Biochem. Soc. Trans. 30, 724-726 ).
  • the present invention describes the detection of pro-hepcidin in the human plasma thereby indicating that hepatocytes secrete the prohormone of hepcidin that may decrease dietary iron absorption via an endocrine pathway. Moreover, hepcidin was detected in HepG2 cells, where the newly discovered transferrin receptor type 2 was also found. The simultaneous existence of hepcidin and TfR2 in HepG2 cells and their common polarized localization and lobular distribution in the liver may indicate that hepcidin is an intrinsic hepatic peptide morphofunctionally coupled to TfR2, which is regulated by transferrin saturation and, in turn, modulates expression of hepcidin. Hence, pertinent findings are expected from studies on the signaling pathway of hepcidin.
  • a Hepcidin enzyme immunoassay (“EIA") is used.
  • EIA is a solid phase enzyme-linked immunosorbent assay (ELISA) based on the competitive principle.
  • ELISA enzyme-linked immunosorbent assay
  • Microtiter wells of a 96 well microtiter plate are coated with a monoclonal mouse anti-Hepcidin antibody.
  • An unknown amount of Hepcidin present in the sample and a fixed amount of Hepcidin conjugated with a biotin molecule compete for the binding sites of the Hepcidin antibodies immobilized on the wells.
  • the microtiter plate is washed to stop the competition reaction. In the following incubation the bound biotin molecules are detected with streptavidin horseradish peroxidase.
  • the plate After one half hour of incubation the plate is washed a second time.
  • the amount of bound biotin conjugate is reverse proportional to the concentration of Hepcidin in the sample.
  • the intensity of colour developed is also reverse proportional to the concentration of Hepcidin in the patient sample.
  • microtiter wells When stored at 2°to 8°C. unbroken reagents will retain reactivity until expiration date. Do not use reagents beyond this date. Microtiter wells must be stored at 2° to 8°C. Once the foilbag has been broken care should be taken to close it tightly again. The immuno-reactivity of the coated microtiter wells is stable for approximately 6 weeks in the broken, but tightly closed plastic zip pouch containing the desiccant.
  • Human or animal serum or EDTA- Heparin- Citrate- plasma or urine should be used in the assay. No special pretreatment of the biological sample is necessary.
  • the biological sample may be stored at 2-8°C. for up to 24 hours, and should be frozen at - 20°C. or lower for longer periods. Do not use grossly hemolyzed or grossly lipemic specimens. For other sample material a special extraction protocol may be necessary.
  • the specimens can be diluted with 0.9 % NaCl and reassayed as described in Assay Procedure.
  • Wash Solution Add deionized water to the 40 x concentrated Wash Solution (contents: 30 ml) to a final volume of 1200 ml.
  • the diluted Wash Solution is stable for 2 weeks at room temperature.
  • Biotin Conjugate Dilute 1 ml of concentrated conjugate solution with 10 ml of conjugate diluent to a final volume of 11 ml.
  • the diluted conjugate is stable for 2 days at 2°- 8°C
  • Testosterone value of each sample is obtained as follows:
  • a DRG ELIZA MAT 3000 and the DRG Regression Program allow the reading and computer assisted interpretation using a four parameter logistic function.
  • the immunization peptide was coupled to keyhole limpet hemocyanin using M-maleimidobenzoyl- N -hydroxysuccinimide ester, and mice were immunized with the peptide conjugate.
  • the peptide epitopes used for the generation of the antibodies displayed no homology to any hitherto reported protein except hepcidin as confirmed by the blast P2 search.
  • FIG. 9A-D RT-PCR analyses using specific primers revealed that hepcidin is highly expressed in human and rat pancreas.
  • the amplification yielded an expected transcript of 201 bp ( FIG. 9A ).
  • Sequence analyses of the amplification products confirmed complete homology with the hepcidin cDNAs of rat and man.
  • the expression of hepcidin in the pancreas was about 13-18% compared to that in the liver ( FIG. 9D ).
  • the existence of hepcidin at the translational level was verified by immunoblotting analyses in human and rat pancreatic extracts. Both N- ( FIG. 9B ) and C-terminal region-specific hepcidin antisera ( FIG.
  • RNA isolation was performed using QIAGEN RNAEasy kit including DNA digestion.
  • reverse transcription (RT)-PCR analysis was performed using the following primers and specification given in 5'-3' orientation: human hepcidin (Genbank database accession no. NM021175), 5'-CCT GAC CAG TGG CTC TGT TT-3'(SEQ ID NO:14) and 5'-GGT TCT ACG TCT TGC AGC AC-3'(SEQ ID NO:15); corresponding to positions 130-149 and 330-311; rat hepcidin (Genbank database accession no.
  • HEPCIDIN IS CO-LOCALIZED WITH INSULIN IN SECRETORY GRANULES OF THE HUMAN AND RAT PANCREATIC ß-CELLS
  • hepcidin antisera under study coincidently immunostained distinct cells within the human Langerhans islets which by their typical location within the islets and by co-localization with insulin were clearly identified as ß-cells ( FIGS. 10A-C and 11A-D ).
  • Other endocrine cell types i.e., glucagon ⁇ -, somatostatin ⁇ -, and pancreatic polypeptide PP-cells
  • the exocrine gland cells were completely unreactive for hepcidin.
  • the same cellular distributive pattern was found in the rat pancreas where strong hepcidin immunoreactivity was detected in ß-cells.
  • hepcidin was confined to the typical secretory granules of the ß-cells, where insulin is localized ( FIG. 12A-B ). Hepcidin immunoreactivity exhibited a distinct intragranular topology; the immunoreactivity for this peptide was absent in the electron-lucent halo but was confined to the dense core of the ß-cell secretory granules, i.e., the compartment where insulin is also resident. Based on this granular co-segregation we assume that hepcidin is co-released with insulin upon glucose stimulation of ß-cells.
  • ß-cells are not only involved in glucose homeostasis but may also regulate iron metabolism by secreting hepcidin and thus exert a dual function. Thereafter, we addressed the question of regulation of hepcidin expression and secretion by glucose and iron.
  • RINM5F cells rat insulinoma cells of the pancreas, exhibiting preserved ß-cell features represent a suitable model for ß-cell experiments.
  • RINM5F cells were grown in RPMI1640 media (PAA, Austria) containing L-glutamine, supplemented with 10% (vol/vol) heat inactivated fetal calf serum, penicillin (100 units/ml) and streptomycin (100 ⁇ g/mL). Cells were cultivated at 37°C in 5% CO 2 . Cells were incubated with increasing concentrations of FENTA from 1 ⁇ M up to 65 ⁇ M for 72 hours and with 100 mM glucose (Merck, Darstadt, Germany) for up to 8 hours. After stimulation experiments, total rna or proteins were extracted from RINM5F cells and analyzed by quantitative PCR and immunoblot assays.
  • RT-PCR analyses using the appropriate primer specifications and combinations successfully employed in the rat liver and pancreas identified expression of hepcidin in RINM5F cells ( FIG.9A-D ).
  • the amplification product in RINM5F cells was also of 201 bp which after sequencing revealed a complete homology to the rat hepcidin cDNA.
  • the existence of the respective translated proteins in these cells was verified by immunocytochemistry and immunoblotting experiments using the region-specific hepcidin antisera (see FIGS. 13A-C and 14A-B ).
  • hepcidin expression in RINm5F cells was analyzed upon exposure to iron, since iron regulates hepcidin expression in the liver.
  • the regulatory effect of iron on hepcidin gene expression was measured in these cells under various FENTA concentrations using quantitative RT-PCR as described, for example, in Gehrke et al., Blood 102: 371-376 (2003 ).
  • a 2-step reverse transcriptase-polymerase chain reaction (RT-PCR) was performed using the Lightcycler system and relative quantification software version 1.0 (ROCHE Molecular Biochemicals; Mannheim, Germany). In a first step, cDNA synthesis was performed with the first strand cDNA synthesis kit for RT-PCR (ROCHE Molecular Biochemicals) according to manufacturer's instructions.
  • transcripts of hepcidin were amplified in duplicates with specific sense and antisense primers. All transcripts were detected using SYBR GREEN I according to manufacturer's instructions and were normalized to ß-actin as internal control.
  • RINM5F cells To analyze whether the experimental findings at the level of RINM5F cells are transferable to in vivo physiological regulatory pathways an oral glucose tolerance test in humans was performed to induce hepcidin secretion by increasing serum glucose levels. Healthy 10 persons started the test in a fasting state (having no food or drink for 12 hours). Initial blood sugar, insulin, and hepcidin were drawn and then the persons were given a glucose solution with 37.5 grams of glucose. The parameters were tested again 30 minutes and 1 hour after drinking the high glucose drink. For hepcidin measurements the hepcidin ELISA competitive binding assay was used. For that assay, blood samples were withdrawn into serum-tubes and centrifuged at 2,500 X G for 10 min at 4°C.
  • the measurements were performed by a hepcidin ELISA competitive binding assay using the antibody EG(2)-Hepn against hepcidin-(28-47), as disclosed, for example, in Kulaksiz, et al. Gut. 53: 735-743 (2004 ), Kulaksiz et al., J. Endocrinol. 184: 361-370 (2005 ) or Pietrangelo, et al., Gastroenterology. 128: 470-479 (2005 ).
  • Determinations were performed in duplicate using 96-well-microtiter plates coated with 200 ⁇ l/well rabbit anti-hepcidin antibody EG(2)-Hepn diluted 1:4000 in TRIS buffered saline (TBS) containing 40 mM TRIS-HCL (PH 7.3), 100 mM NaCl. 50 ⁇ l standards containing various amounts of synthetic peptides (0, 20, 100, 500, and 1000 ng/ml) or human serum samples and 150 ⁇ L N-terminally biotinylated hepcidin-(28-47) (Peptide Specialty Laboratories GMBH, Heidelberg, Germany) (2 ng/well) were added to each well and incubated for 1 hour at RT.
  • TRIS buffered saline TRIS buffered saline
  • PH 7.3 40 mM TRIS-HCL
  • 50 ⁇ l standards containing various amounts of synthetic peptides (0, 20, 100, 500, and 1000 ng/ml) or human
  • the biotinylated antigen-antibody complexes were detected by streptavidin-peroxidase enzyme (DAKO, Hamburg, Germany) with the substrate tetramethylbenzidine (DRG INSTRUMENTS GMBH, Marburg, Germany); the color reaction was stopped with 1 m H 2 SO 4 and the extinction of the solution was read at 450 / 630 nm wavelength.
  • RNA isolation was performed using QIAGEN RNAEasy kit including DNA digestion.
  • reverse transcription (RT)-PCR analysis was performed using the following primers and specifications given in 5'-3' orientation: rat ferroportin (Genbank database accession no.
  • cR293 cells were stably transfected with a plasmid containing an ecdysone-regulated Ferroportin-GFP (Fpn-GFP) construct.
  • Fpn-GFP ecdysone-regulated Ferroportin-GFP
  • bioactive hepcidin binds to ferroportin and specifically induces the internalization of Fpn-GFP.
  • Fpn-GFP was induced by ponasterone treatment for 24 h. Cells were then incubated with 0.1 to 0.5 ⁇ M purified human hepcidin (from urine) for 6 h and imaged by epifluorescence microscopy.
  • the invention provides sensitive diagnostic methods and kits enabling the detection of hepcidin in human plasma, urine and other body fluids.
  • the invention opens a broad range of therapeutic perspectives, where a hepcidin antibody and diagnostic methods and kits can be used for the determination of hepcidin as a parameter for the progress of the diseases mentioned above during and after therapy.
  • the inventors of the current invention were successful after diligent testing to establish a reliable ELISA specific to the C-terminal of hepcidin.
  • the following examples demonstrate the difficulty of establishing a reliable ELISA specific to the C-terminal of hepcidin.
  • microtiter plate was coated with anti-hepcidin antibodies and incubated with biotinylated Hepcidin-25 peptide. Streptavidin HRP and TMB were used for detection of the bound hepcidin conjugates. No binding occurred.
  • the activated microtiter plate (protein immobilizer) was, via covalent binding, coated with Hepcidin-25.
  • the conjugate biotinylated hepcidin
  • the conjugate was provided at different concentrations and simoultaneously incubated with rabbit anti-hepcidin antibodies. Bound antibodies were detected with goat anti-rabbit HRP conjugate TMB. Suppression of the antibody binding to antigen was not detected (i.e. no difference in the measured optical densities).
  • Biotinylated Hepcidin-25 was immobilized on a streptavidin coded plate. A simultaneous incubation with rabit anti Hepcidin-25 followed and the following Hepcidin-preparations and probes were used:
  • Recombinant Hepcidin molecule was immobilized, via His-tag, on a Ni-chelate plate. Incubation with rabit anti Hepcidin antibodies and differing dilution steps followed with subsequent detection of antibody binding with goat anti rabit HRP conjugate and TMB substrate. The binding of antibodies on the immobilized Hepcidin-25 reflects the respective dilution steps. However, a standard curve was not able to be established.
  • Experiment 5 Attempt of a suppressing the antigen from binding (follow-up experiment to above-mentioned Experiment 4).
  • Plate coating was analogous to Experiments 4 and 5.
  • the serum samples were treated with immobilized TCEP (Pierce) to reduce the hepcidin molecule in the samples and therewith modify the structure so that they would be recognized by the antibodies.
  • Simultaneous incubation of the rabbit anti Hepcidin antibody with the reduced serum samples was performed.
  • the detection of binding was performed using goat anti rabit HRP conjugates and TMB substrate. No suppression of antigens/antibody binding was observed.
  • Biotinylated Hepcidin-25 was immobilized on a microtiter plate coated with streptavidin. A following simultaneous incubation of monoclonal antibodies with Hepcidin-25 in differing dilution steps followed. Detection of binding was performed with goat anti-mouse HRP conjugate and TMB substrate. Suppression of the immobilized antigen via the addition of free hepcidin was detected. However, a standard curve was not able to be established.
  • Mouse anti hepcidin antibodies (mHK(9)) were immobilized on a microtiter plate with goat anti mouse antibodies. A simultaneous incubation of biotinylated Hepcidin-25 conjugate followed with an unmarked hepcidin molecule from the standard and native Hepcidin-25 from serum samples and plasma samples. Detection of binding was performed with Streptavidin HRP conjugate and TMB. A sensitive quantitative assay for the detection of Hepcidin-25 in serum samples as well as plasma samples was established, as indicated in Figures 17 and 18 .
  • MONOCLONAL ANTIBODIES WHICH BIND SEQ. ID. NO. 3 ARE SUITABLE FOR USE IN ELISA FOR THE MATURE 20-25 AMINO ACID LONG FORM OF HEPCIDINHepcidin-25 ELISA
  • the coated microtiterplate is stabilized with phosphate buffer pH 7.2 containing sucrose and BSA.
  • the invention has applications in connection with diagnosing a disease condition characterized by non-physiological levels of hepcidin protein, including prohepcidin and fragments thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Endocrinology (AREA)
  • General Health & Medical Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Organic Chemistry (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Claims (38)

  1. Anticorps isolé qui se lie à une séquence d'acides aminés comprenant la SEQ ID NO : 4 qui est hrskxgmc, où ledit anticorps se lie spécifiquement à la SEQ. ID. NO. 4, où x dans la SEQ ID NO : 4 est un acide aminé quelconque choisi dans le groupe constitué des formes stéréoisomères L ou D des 20 acides aminés communs, ainsi que de l'acide aminé acide alpha aminobutyrique, et où ledit anticorps détecte l'hepcidine mature longue de 20-25 acides aminés et se lie à celle-ci.
  2. Anticorps selon la revendication 1, dans lequel le X de la SEQ ID NO : 4 est l'acide alpha aminobutyrique comme indiqué dans la SEQ ID NO : 5.
  3. Anticorps selon la revendication 1, dans lequel le X de la SEQ ID NO : 4 est la cystéine comme indiqué dans la SEQ ID NO : 3.
  4. Anticorps selon l'une quelconque des revendications 1 à 3, dans lequel ledit anticorps se lie à un ou plusieurs épitopes dans ladite SEQ ID NO.
  5. Anticorps selon l'une quelconque des revendications 1 à 4, dans lequel ledit anticorps est un anticorps monoclonal.
  6. Anticorps selon l'une quelconque des revendications 1 à 4, dans lequel ledit anticorps est un anticorps polyclonal.
  7. Anticorps selon l'une quelconque des revendications 1 à 5, dans lequel ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
  8. Anticorps selon la revendication 5, dans lequel l'anticorps est un anticorps monoclonal humanisé.
  9. Anticorps selon la revendication 5, dans lequel l'anticorps est un anticorps monoclonal humain.
  10. Séquence d'acides aminés isolée qui est la SEQ ID NO : 4 qui est hrskxgmc, où x dans la SEQ. ID. NO. 4 est un acide aminé quelconque choisi dans le groupe constitué des formes stéréoisomères L ou D des 20 acides aminés communs, ainsi que de l'acide aminé acide alpha aminobutyrique.
  11. Séquence d'acides aminés isolée de la revendication 10, dans laquelle le X de la SEQ ID NO : 4 est un acide alpha aminobutyrique comme indiqué dans la SEQ ID NO : 5.
  12. Séquence d'acides aminés isolée de la revendication 10, dans laquelle le X de la SEQ ID NO : 4 est la cystéine comme indiqué dans la SEQ ID NO : 3.
  13. Procédé de production d'un anticorps monoclonal qui se lie à une séquence d'acides aminés comprenant la SEQ ID NO : 4 qui est hrskxgmc, où ledit anticorps se lie spécifiquement à la SEQ ID NO : 4, comprenant le fait : d'immuniser un mammifère non-humain avec la séquence d'acides aminés de la SEQ ID NO : 5 ; de récolter des cellules B dudit mammifère ; de créer des hybridomes à partir des cellules B récoltées, où lesdits hybridomes produisent des anticorps ; et de sélectionner des hybridomes qui produisent des anticorps se liant spécifiquement à la SEQ ID NO : 4, où x dans la SEQ ID NO : 4 est un acide aminé quelconque choisi dans le groupe constitué des formes stéréoisomères L ou D des 20 acides aminés communs, ainsi que de l'acide aminé acide alpha aminobutyrique.
  14. Procédé de la revendication 13, dans lequel X de la SEQ ID NO : 4 est l'acide alpha aminobutyrique comme indiqué dans la SEQ ID NO : 5.
  15. Procédé de la revendication 13, dans lequel X de la SEQ ID. NO : 4 est la cystéine comme indiqué dans la SEQ ID NO : 3.
  16. Procédé de l'une quelconque des revendications 13 à 15, dans lequel ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
  17. Procédé permettant de détecter et de quantifier la quantité d'une protéine hepcidine dans un échantillon de tissu ou de fluide, comprenant le fait d'obtenir l'échantillon de tissu ou de fluide à partir d'un sujet ; de mettre en contact l'échantillon de tissu ou de fluide in vitro avec un anticorps ou un fragment de celui-ci qui se lie à une séquence d'acides aminés comprenant la SEQ ID NO : 4 qui est hrskxgmc, où ledit anticorps se lie spécifiquement à la SEQ ID NO : 4, où x dans la SEQ ID NO : 4 est un acide aminé quelconque choisi dans le groupe constitué des formes stéréoisomères L ou D des 20 acides aminés communs, ainsi que de l'acide aminé acide alpha aminobutyrique, et où ledit anticorps détecte l'hepcidine mature longue de 20-25 acides aminés et se lie à celle-ci, et de détecter et de quantifier le niveau d'hepcidine dans l'échantillon, où un niveau non physiologique de la protéine hepcidine indique un état pathologique.
  18. Procédé de la revendication 17, dans lequel le X de la SEQ ID NO : 4 est l'acide alpha aminobutyrique comme indiqué dans la SEQ ID NO : 5.
  19. Procédé de la revendication 17, dans lequel X de la SEQ ID NO : 4 est la cystéine comme indiqué dans la SEQ ID NO : 3.
  20. Procédé de l'une quelconque des revendications 17 à 19, dans lequel ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
  21. Procédé de l'une quelconque des revendications 17 à 20, dans lequel le procédé comprend un deuxième anticorps qui se lie spécifiquement à la séquence d'acides aminés d'hepcidine entre les acides aminés 28-47 comme indiqué dans la SEQ ID NO : 8, qui représente la pro-hepcidine ; et la détection et la quantification du niveau de la pro-hepcidine dans l'échantillon ; et la comparaison des valeurs de la pro-hepcidine aux valeurs obtenues dans l'une quelconque des revendications 17 à 20 pour déterminer le niveau de l'hepcidine mature dans l'échantillon.
  22. Procédé de l'une quelconque des revendications 17 à 21, dans lequel le procédé de détection est la Méthode Immuno-enzymatique, abrégée par ELISA.
  23. Procédé de l'une quelconque des revendications 17 à 22, dans lequel ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
  24. Kit permettant de détecter un état pathologique caractérisé par des niveaux non physiologiques de la pro-hepcidine ou de l'hepcidine mature, comprenant un anticorps selon l'une quelconque des revendications 1 à 9, et un réactif qui se lie directement ou indirectement audit anticorps ou à son fragment.
  25. Kit de la revendication 24, dans lequel le kit comprend en outre un deuxième anticorps ou un fragment de celui-ci, capable de se lier spécifiquement à la SEQ ID NO : 8 ; et un réactif qui se lie, directement ou indirectement, audit anticorps ou à son fragment.
  26. Kit de la revendication 24 ou 25, dans lequel l'anticorps ou son fragment est immobilisé sur un support.
  27. Kit de l'une quelconque des revendications 24 à 26, dans lequel le réactif comprend la pro-hepcidine ou l'hepcidine mature complexée avec une première molécule de liaison.
  28. Kit de l'une quelconque des revendications 24 à 27, dans lequel l'état pathologique est choisi dans le groupe constitué de perturbations du métabolisme du fer : surcharge ou carence en fer primaire ou secondaire, telle que l'anémie ferriprive, l'anémie d'une maladie chronique, des maladies de surcharge en fer génétique et nongénétique, telles que l'hémosidérose et l'hémochromatose ou l'hémochromatose secondaire, l'acéruloplasminémie, l'hypotransferrinémie, l'atransferrinémie ; la thalassémie, des maladies du foie provoquées par l'alcool, des inflammations et des infections, et notamment la sepsie ; l'anémie dans la maladie de Crohn, l'anémie dans la recto-colite hémorragique, et une combinaison quelconque de celles-ci.
  29. Kit de l'une quelconque des revendications 24 à 28, dans lequel ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
  30. Composition pharmaceutique comprenant l'un quelconque des anticorps des revendications 1 à 9 pour l'utilisation en tant que médicament.
  31. Composition pharmaceutique comprenant l'un quelconque des anticorps des revendications 1 à 9, pour une utilisation dans un procédé de traitement d'un état pathologique choisi dans le groupe constitué de carence en fer primaire ou secondaire, telle que l'anémie ferriprive, l'anémie d'une maladie chronique, des inflammations et des infections, et notamment la sepsie ; l'anémie dans la maladie de Crohn, l'anémie dans la recto-colite hémorragique, et une combinaison quelconque de celles-ci.
  32. Composition pharmaceutique pour une utilisation selon la revendications 30 ou 31, dans laquelle ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
  33. Procédé de surveillance d'un état pathologique caractérisé par des niveaux non physiologiques de l'hepcidine chez un sujet, comprenant le fait de détecter et de quantifier une quantité de la protéine hepcidine ou du précurseur de l'hepcidine à un premier temps in vitro, de détecter et de quantifier une quantité de la protéine hepcidine ou du précurseur de l'hepcidine à un deuxième temps ultérieur in vitro, moyennant quoi |H1 - N|<|H2 - N| indique que l'état pathologique a progressé, et |H1 - N|>|H2 - N| indique que l'état pathologique s'est amélioré, où H1 est égal à la quantité de la protéine hepcidine ou du précurseur de l'hepcidine mesurée au premier temps, H2 est égal à la quantité de la protéine hepcidine ou du précurseur de l'hepcidine mesurée au deuxième temps, et N est égal à la plage ou quantité normale de la protéine hepcidine ou du précurseur de l'hepcidine ; où la détection et la quantification sont conformes à la revendication 17.
  34. Procédé de la revendication 33, dans lequel le sujet a été traité de manière thérapeutique pour l'état pathologique.
  35. Procédé de la revendication 33 ou 34, dans lequel l'état pathologique est choisi dans le groupe constitué de perturbations du métabolisme du fer, telle qu'une surcharge ou une carence en fer primaire ou secondaire, telle que l'anémie ferriprive, l'anémie d'une maladie chronique, des maladies de surcharge en fer génétique et nongénétique, telles que l'hémosidérose et l'hémochromatose ou l'hémochromatose secondaire, l'acéruloplasminémie, l'hypotransferrinémie, l'atransferrinémie ; la thalassémie, des maladies du foie provoquées par l'alcool, des inflammations et des infections, et notamment la sepsie ; l'anémie dans la maladie de Crohn, l'anémie dans la recto-colite hémorragique, et une combinaison quelconque de celles-ci.
  36. Procédé de l'une quelconque des revendications 33 à 35, dans lequel le sujet a été traité de manière thérapeutique avec substitution de fer par voie orale ou intraveineuse.
  37. Procédé de l'une quelconque des revendications 33 à 35, dans lequel le sujet a été traité de manière thérapeutique avec une thérapie à base d'érytropoïétine.
  38. Procédé de l'une quelconque des revendications 33 à 35, dans lequel ledit anticorps est mHk (9) déposé sous le numéro d'accès DSM ACC2812.
EP07723512.5A 2007-01-25 2007-03-22 Procede de diagnostic de maladies par depistage de l'hepcidine dans des tissus, du sang ou des liquides organiques humains ou animaux, anticorps monoclonaux specifiques de l'hepcidine humaine et leurs utilisations associees Not-in-force EP2109624B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/657,772 US8017737B2 (en) 2002-11-19 2007-01-25 Diagnostic methods for diseases by screening for hepcidin in human or animal tissues, blood or body fluids; monoclonal antibodies specific to human hepcidin and associated uses therefor
PCT/EP2007/002559 WO2008089795A1 (fr) 2007-01-25 2007-03-22 Procédé de diagnostic de maladies par dépistage de l'hepcidine dans des tissus, du sang ou des liquides organiques humains ou animaux, anticorps monoclonaux spécifiques de l'hepcidine humaine et leurs utilisations associées

Publications (2)

Publication Number Publication Date
EP2109624A1 EP2109624A1 (fr) 2009-10-21
EP2109624B1 true EP2109624B1 (fr) 2014-07-16

Family

ID=39645313

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07723512.5A Not-in-force EP2109624B1 (fr) 2007-01-25 2007-03-22 Procede de diagnostic de maladies par depistage de l'hepcidine dans des tissus, du sang ou des liquides organiques humains ou animaux, anticorps monoclonaux specifiques de l'hepcidine humaine et leurs utilisations associees

Country Status (3)

Country Link
US (2) US8017737B2 (fr)
EP (1) EP2109624B1 (fr)
WO (1) WO2008089795A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9657098B2 (en) 2013-03-15 2017-05-23 Intrinsic Lifesciences, Llc Anti-hepcidin antibodies and uses thereof
US10323088B2 (en) 2014-09-22 2019-06-18 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4944360B2 (ja) 2001-05-25 2012-05-30 アンスティテュ ナシオナル ド ラ サント エ ド ラ ルシュルシェ メディカル(アンセルム) 鉄ホメオスタシスの障害を治療するための医薬品を製造するためのヘプシジンの使用
US8017737B2 (en) 2002-11-19 2011-09-13 Hasan Kulaksiz Diagnostic methods for diseases by screening for hepcidin in human or animal tissues, blood or body fluids; monoclonal antibodies specific to human hepcidin and associated uses therefor
EP2016177A2 (fr) * 2006-04-12 2009-01-21 Isis Pharmaceuticals, Inc. Compositions et leurs utilisations sur l'hepcidine
CA2663581C (fr) 2006-09-21 2016-03-01 Alnylam Pharmaceuticals, Inc. Compositions et procedes servant a inhiber l'expression du gene hamp
US8629250B2 (en) * 2007-02-02 2014-01-14 Amgen Inc. Hepcidin, hepcidin antagonists and methods of use
ES2385772T3 (es) * 2007-10-02 2012-07-31 Institut National De La Sante Et De La Recherche Medicale (Inserm) Anticuerpos específicos para la hepcidina humana
WO2009055078A2 (fr) * 2007-10-26 2009-04-30 University Of Utah Research Foundation Identification du site de liaison de l'hepcidine sur la ferroportine
CL2008003190A1 (es) * 2007-11-02 2009-09-04 Lilly Co Eli Anticuerpo que se enlaza selectivamente a hepcidina-25 humana madura; polinucleotido codificante, vector y celula huesped que lo comprende; uso medico para tratar la anemia, incrementar nivel de hierro, conteo de rediculocito, celulas rojas, hemoglobina, o hematocritos; proceso de produccion; composicion farmaceutica.
WO2009139822A1 (fr) * 2008-05-01 2009-11-19 Amgen Inc. Anticorps anti-hepcidine et méthodes d’utilisation associées
CA2733497C (fr) 2008-08-06 2015-06-02 Eli Lilly And Company Anticorps selectifs anti-hepcidine-25 et leurs utilisations
DE102009034150A1 (de) * 2009-07-20 2011-01-27 Fresenius Medical Care Deutschland Gmbh Adsorbens zur Adsorption von Hepcidin
WO2011071368A1 (fr) 2009-12-11 2011-06-16 Umc St. Radboud Méthode de mesure de l'hepcidine
WO2011143232A1 (fr) * 2010-05-10 2011-11-17 Westerman Mark E Marqueurs de lésion rénale aiguë
EP2723861A4 (fr) 2011-06-21 2014-12-10 Alnylam Pharmaceuticals Inc Compositions et procédés d'inhibition de l'expression du peptide antimicrobien hepcidine (hamp) ou du gène lié à hamp
CN103123355B (zh) * 2012-12-28 2015-04-15 常州市第一人民医院 一种用于前列腺癌骨转移诊断的试剂盒及其使用方法
US10947543B2 (en) * 2013-03-13 2021-03-16 Yale University Interferon production using short RNA duplexes
CN112345772A (zh) * 2020-10-29 2021-02-09 江苏沃兴生物科技有限公司 一种基于单克隆抗体制备的铁调素的胶乳增强比浊法检测试剂盒
US11649457B2 (en) 2020-12-09 2023-05-16 Yale University Methods for treating SARS-CoV-2 infection
US11786545B2 (en) 2020-12-09 2023-10-17 Yale University Compositions and methods for treating SARS-CoV-2 infection

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4464465A (en) * 1982-04-05 1984-08-07 Genetic Systems Corporation Cell-driven viral transfer in eukaryotes
US5314804A (en) 1992-03-24 1994-05-24 Serim Research Corporation Test for Helicobacter pylori
US5753230A (en) * 1994-03-18 1998-05-19 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
EP1179060A4 (fr) 1999-05-13 2003-03-26 Human Genome Sciences Inc 143 proteines humaines secretees
EP1210418B1 (fr) 1999-06-02 2010-08-18 Genentech, Inc. Polypeptides secretes et transmembranaires ainsi que les acides nucleiques codant pour ceux-ci
US6262247B1 (en) 1999-08-30 2001-07-17 Incyte Genomics, Inc. Polycyclic aromatic hydrocarbon induced molecules
JP4944360B2 (ja) * 2001-05-25 2012-05-30 アンスティテュ ナシオナル ド ラ サント エ ド ラ ルシュルシェ メディカル(アンセルム) 鉄ホメオスタシスの障害を治療するための医薬品を製造するためのヘプシジンの使用
GB0116047D0 (en) 2001-06-29 2001-08-22 Implyx Ltd Peptide motif for therapy
EP1578254B1 (fr) * 2002-11-19 2013-03-06 DRG International, Inc. Methode diagnostique pour maladies par criblage d'hepcidine dans des tissus, du sang ou des fluides corporels humains ou animaux et utilisations therapeutiques
US8017737B2 (en) 2002-11-19 2011-09-13 Hasan Kulaksiz Diagnostic methods for diseases by screening for hepcidin in human or animal tissues, blood or body fluids; monoclonal antibodies specific to human hepcidin and associated uses therefor
US7411048B2 (en) 2002-11-19 2008-08-12 Drg International, Inc. Diagnostic method for diseases by screening for hepcidin in human or animal tissues, blood or body fluids

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9657098B2 (en) 2013-03-15 2017-05-23 Intrinsic Lifesciences, Llc Anti-hepcidin antibodies and uses thereof
US9803011B2 (en) 2013-03-15 2017-10-31 Intrinsic Lifesciences Llc Anti-hepcidin antibodies and uses thereof
US10239941B2 (en) 2013-03-15 2019-03-26 Intrinsic Lifesciences Llc Anti-hepcidin antibodies and uses thereof
US10323088B2 (en) 2014-09-22 2019-06-18 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof

Also Published As

Publication number Publication date
US8304258B2 (en) 2012-11-06
EP2109624A1 (fr) 2009-10-21
US20070224186A1 (en) 2007-09-27
US8017737B2 (en) 2011-09-13
US20120003696A1 (en) 2012-01-05
WO2008089795A1 (fr) 2008-07-31

Similar Documents

Publication Publication Date Title
EP2109624B1 (fr) Procede de diagnostic de maladies par depistage de l&#39;hepcidine dans des tissus, du sang ou des liquides organiques humains ou animaux, anticorps monoclonaux specifiques de l&#39;hepcidine humaine et leurs utilisations associees
US7998691B2 (en) Diagnostic method for diseases by screening for hepcidin in human or animal tissues, blood or body fluids and therapeutic uses therefor
US8003338B2 (en) Diagnostic method for diseases by screening for hepcidin in human or animal tissues, blood or body fluids and therapeutic uses therefor
US9304127B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment for use in therapy
EP2594588B1 (fr) Anticorps anti-adrénomédulline (ADM) ou fragment d&#39;anticorps anti-ADM ou échafaudage protéique non-Ig anti-ADM destinés à être utilisés en thérapie
CN105102985B (zh) 用于指导血压下降疗法的肾上腺髓质素
EP2780371B1 (fr) Anticorps anti-adrénomédulline (adm) ou fragment d&#39;anticorps anti-adm ou échafaudage non-ig anti-adm pour la régulation de l&#39;équilibre de fluide chez un patient atteint d&#39;une maladie chronique ou aiguë
JP2019528676A (ja) 抗amh抗体を調製するための方法及びその使用
JP6737592B2 (ja) 抗活性型gip抗体
CN102232087A (zh) 修饰的人igf-1/e肽的抗体
KR100493932B1 (ko) 레지스틴에 대한 단클론 항체, 이의 제조 방법, 및 용도
JP6586648B2 (ja) 抗プログルカゴン抗体
JP4851620B2 (ja) Nk4に対するモノクローナル抗体
JP2011160696A (ja) 修飾ヒトigf−1/eペプチドに対する抗体
JPH0523196A (ja) ヒトグリセンチンのモノクローナル抗体およびそれを用いるヒトグリセンチンの定量法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090825

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

17Q First examination report despatched

Effective date: 20091117

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602007037644

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C07K0016260000

Ipc: A61K0039395000

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 39/395 20060101AFI20120912BHEP

Ipc: C07K 16/26 20060101ALI20120912BHEP

Ipc: G01N 33/74 20060101ALI20120912BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20130502

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20131004

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140224

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 677157

Country of ref document: AT

Kind code of ref document: T

Effective date: 20140815

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602007037644

Country of ref document: DE

Effective date: 20140828

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: E. BLUM AND CO. AG PATENT- UND MARKENANWAELTE , CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20140716

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 677157

Country of ref document: AT

Kind code of ref document: T

Effective date: 20140716

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141016

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141117

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141116

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602007037644

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20150417

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150322

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20070322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140716

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20180308

Year of fee payment: 12

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190322

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20210427

Year of fee payment: 15

Ref country code: FR

Payment date: 20210427

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20210415

Year of fee payment: 15

Ref country code: CH

Payment date: 20210423

Year of fee payment: 15

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602007037644

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20220322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220331

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220322

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220331

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221001

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220331